EP2655418A2 - Anticorps et immunoconjugués anti-mésothéline - Google Patents

Anticorps et immunoconjugués anti-mésothéline

Info

Publication number
EP2655418A2
EP2655418A2 EP11810734.1A EP11810734A EP2655418A2 EP 2655418 A2 EP2655418 A2 EP 2655418A2 EP 11810734 A EP11810734 A EP 11810734A EP 2655418 A2 EP2655418 A2 EP 2655418A2
Authority
EP
European Patent Office
Prior art keywords
antibody
seq
mesothelin
hvr
amino acid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
EP11810734.1A
Other languages
German (de)
English (en)
Other versions
EP2655418B1 (fr
Inventor
Mark Dennis
Suzanna J. Scales
Susan D. Spencer
Yin Zhang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
F Hoffmann La Roche AG
Original Assignee
F Hoffmann La Roche AG
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=45498116&utm_source=***_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=EP2655418(A2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by F Hoffmann La Roche AG filed Critical F Hoffmann La Roche AG
Priority to EP17194505.8A priority Critical patent/EP3296321B1/fr
Publication of EP2655418A2 publication Critical patent/EP2655418A2/fr
Application granted granted Critical
Publication of EP2655418B1 publication Critical patent/EP2655418B1/fr
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/08Peptides having 5 to 11 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/6811Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a protein or peptide, e.g. transferrin or bleomycin
    • A61K47/6817Toxins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • A61K47/6869Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell the tumour determinant being from a cell of the reproductive system: ovaria, uterus, testes, prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1027Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against receptors, cell-surface antigens or cell-surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57423Specifically defined cancers of lung
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57438Specifically defined cancers of liver, pancreas or kidney
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57442Specifically defined cancers of the uterus and endometrial
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57449Specifically defined cancers of ovaries
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/567Framework region [FR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/77Internalization into the cell
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants

Definitions

  • the present invention relates to anti-mesothelin antibodies and immunoconjugates and methods of using the same.
  • Mesothelin is a cell surface glycoprotein with expression normally restricted to mesothelia (peritoneum, pericardium, and pleura). However, mesothelin is significantly overexpressed in a variety of tumor types. Mesothelin interacts with MUC16 (also called CA125), a mucin-like glycoprotein previously identified as an ovarian tumor antigen. MUC16 has an extracellular domain comprising at least 14,000 residues and characterized by tandem repeats of 156 amino acids each, referred to as mucin repeats. (See, e.g., O'Brien et al, Tumour Biol. 22:348-366 (2001); Yin et al, J. Biol. Chem.
  • Mesothelin is synthesized as a 71 kDa precursor protein, the mature portion of which is expressed on the cell surface. That precursor protein is proteolytically cleaved by furin into a 31 kDa shed component (referred to as megakaryocyte potentiating factor, or MPF) and a 40 kDa mesothelin component. The latter component may remain associated with the cell surface via a GPI linkage but may also be shed through a proteolytic mechanism.
  • MPF megakaryocyte potentiating factor
  • the latter component may remain associated with the cell surface via a GPI linkage but may also be shed through a proteolytic mechanism.
  • the invention provides anti-mesothelin antibodies and immunoconjugates and methods of using the same.
  • an isolated antibody that binds to mesothelin wherein the antibody is selected from: (i) an antibody that binds an epitope of SEQ ID NO:43 comprising El 53 and D174 and that optionally has one or more of the following characteristics: (a) does not exhibit reduced binding to glycosylated forms of mesothelin; (b) does not block binding of mesothelin to MUC16; and (c) binds mesothelin with an affinity of ⁇ 5 nM; (ii) an antibody that binds an epitope of SEQ ID NO:43 comprising E211 and that optionally has one or more of the following characteristics: (a) does not block binding of mesothelin to MUC16; and (b) binds mesothelin with an affinity of ⁇ 5 nM; and (iii) an antibody that binds to an epitope within amino acids 1-131 of SEQ ID NO :43 and binds mesothel
  • the antibody is a monoclonal antibody. In certain embodiments, the antibody is a human, humanized, or chimeric antibody. In certain embodiments, the antibody is an antibody fragment that binds mesothelin. In certain embodiments, the mesothelin is human mesothelin of SEQ ID NO:43.
  • the antibody comprises: (a) (i) HVR-H3 comprising the amino acid sequence of SEQ ID NO:22, (ii) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 19, and (iii) HVR-H2 comprising the amino acid sequence of SEQ ID NO:21; (b) (i) HVR-H3 comprising the amino acid sequence of SEQ ID NO:39, (ii) HVR-L3 comprising the amino acid sequence of SEQ ID NO:35, and (iii) HVR-H2 comprising the amino acid sequence of SEQ ID NO:37; or (c) HVR-H3, HVR-L3, and HVR-H2 of the antibody produced by hybridoma 19C3 having ATCC Accession No.
  • the antibody comprises (a) (i) HVR-Hl comprising the amino acid sequence of SEQ ID NO:20, (ii) HVR-H2 comprising the amino acid sequence of SEQ ID NO:21, and (iii) HVR-H3
  • HVR-Hl comprising the amino acid sequence of SEQ ID NO:36
  • HVR-H2 comprising the amino acid sequence of SEQ ID NO:37
  • HVR-H3 comprising the amino acid sequence of SEQ ID NO:39
  • the antibody comprises (a) (i) HVR-Hl comprising the amino acid sequence of SEQ ID NO:20, (ii) HVR-H2 comprising the amino acid sequence of SEQ ID NO:21 , (iii) HVR-H3 comprising the amino acid sequence of SEQ ID NO:22, (iv) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 17, (v) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 18, and (vi) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 19; (b) (i) HVR-H1 comprising the amino acid sequence of SEQ ID NO:36, (ii) HVR-H2 comprising the amino acid sequence of SEQ ID NO:37, (iii) HVR-H3 comprising the amino acid sequence of SEQ ID NO:39, (iv) HVR-L1 comprising the amino acid sequence of SEQ ID NO:33, (v) HVR-L2 comprising the amino acid
  • the antibody comprises (i) HVR-H1 comprising the amino acid sequence of SEQ ID NO:20, (ii) HVR-H2 comprising the amino acid sequence of SEQ ID NO:21 , (iii) HVR-H3 comprising the amino acid sequence of SEQ ID NO:22, (iv) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 17, (v) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 18, and (vi) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 19, and further comprising a light chain variable domain comprising a framework FR2 sequence of SEQ ID NO:25 and an FR3 sequence of SEQ ID NO:27.
  • the antibody comprises (a) (i) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 17, (ii) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 18, and (iii) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 19; (b) (i) HVR- Ll comprising the amino acid sequence of SEQ ID NO: 33, (ii) HVR-L2 comprising the amino acid sequence of SEQ ID NO:34, and (iii) HVR-L3 comprising the amino acid sequence of SEQ ID NO:35; or (c) HVR-L1 , HVR-L2 and HVR-L3 of the antibody produced by hybridoma 19C3 having ATCC Accession No.
  • the antibody comprises HVR-L1 comprising the amino acid sequence of SEQ ID NO: 17, HVR-L2 comprising the amino acid sequence of SEQ ID NO: 18, and HVR-L3 comprising the amino acid sequence of SEQ ID NO: 19, and further comprises a light chain variable domain comprising a framework FR2 sequence of SEQ ID NO:25 and an FR3 sequence of SEQ ID NO:27.
  • the antibody comprises (a) a VH sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 8; (b) a VL sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO:4; (c) a VH sequence as in (a) and a VL sequence as in (b); (d) a VH sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 16; (e) a VL sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 12; (f) a VH sequence as in (d) and a VL sequence as in (e); (g) a VH sequence having at least 95% sequence identity to the amino acid sequence of the VH sequence of the antibody produced by hybridoma 19C3 having ATCC Accession No.
  • the antibody comprises a VH sequence of SEQ ID NO: 8, a VH sequence of SEQ ID NO: 16, or a VH sequence of the antibody produced by hybridoma 19C3 having ATCC Accession No. PTA-11464.
  • the antibody comprises a VL sequence of SEQ ID NO:4, a VL sequence of SEQ ID NO: 12, or a VL sequence of the antibody produced by hybridoma 19C3 having ATCC Accession No. PTA-
  • the invention provides an antibody comprising (a) a VH sequence of SEQ ID NO: 8 and a VL sequence of SEQ ID NO:4; (b) a VH sequence of SEQ ID NO: 16 and a VL sequence of SEQ ID NO: 12; (c) a VH sequence and a VL sequence of the antibody produced by hybridoma 19C3 having ATCC Accession No. PTA-11464; or (d) the antibody produced by hybridoma 19C3 having ATCC Accession No. PTA-11464.
  • an antibody according to any of the above embodiments is an IgG 1 , IgG2a or IgG2b antibody.
  • the invention provides an isolated nucleic acid encoding an antibody according to any of the above embodiments.
  • a host cell comprising the nucleic acid is provided.
  • a method of producing an antibody is provided, the method comprising culturing the host cell so that the antibody is produced.
  • Ab is an antibody as in any of the above embodiment
  • R 7 is sec-butyl, each R 8 is independently selected from CH 3 , 0-CH 3 , OH, and H; R 9 is H; and R 18 is -C(R 8 ) 2 -C(R 8 ) 2 -aryl; and
  • the drug is an auristatin. In one such embodiment, the drug is MMAE.
  • the linker is cleavable by a protease. In one such embodiment, the linker comprises a val-cit dipeptide.
  • the immunoconjugate has the formula:
  • the antibody comprises (i) HVR-Hl comprising the amino acid sequence of SEQ ID NO:20, (ii) HVR-H2 comprising the amino acid sequence of SEQ ID NO:21, (iii) HVR-H3 comprising the amino acid sequence of SEQ ID NO:22, (iv) HVR-L1 comprising the amino acid sequence of SEQ ID NO : 17, (v) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 18, and (vi) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 19.
  • the antibody comprises (i) HVR-Hl comprising the amino acid sequence of SEQ ID NO:36, (ii) HVR-H2 comprising the amino acid sequence of SEQ ID NO:37, (iii) HVR-H3 comprising the amino acid sequence of SEQ ID NO:39, (iv) HVR-L1 comprising the amino acid sequence of SEQ ID NO:33, (v) HVR-L2 comprising the amino acid sequence of SEQ ID NO:34, and (vi) HVR-L3 comprising the amino acid sequence of SEQ ID NO:35.
  • the antibody comprises (a) a VH sequence of SEQ ID NO: 8 and a VL sequence of SEQ ID NO:4.
  • the antibody comprises (b) a VH sequence of SEQ ID NO: 16 and a VL sequence of SEQ ID NO: 12.
  • the invention provides a pharmaceutical formulation comprising an immunoconjugate as in any of the above embodiments and a pharmaceutically acceptable carrier.
  • the pharmaceutical formulation further comprises an additional therapeutic agent.
  • the additional therapeutic agent is gemcitabine.
  • the additional therapeutic agent is an anti-MUC16 antibody conjugated to a cytotoxic agent.
  • the invention provides an immunoconjugate as in any of the above embodiments for use as a medicament.
  • the invention provides an immunoconjugate as in any of the above embodiments for use in treating a mesothelin-positive cancer.
  • the mesothelin-positive cancer is selected from pancreatic cancer, ovarian cancer, lung cancer, endometrial cancer, and mesothelioma.
  • the mesothelin-positive cancer is a dual-positive cancer.
  • the invention provides for use of an immunoconjugate as in any of the above embodiments in the manufacture of a medicament.
  • the medicament is for treatment of a mesothelin-positive cancer.
  • the mesothelin-positive cancer is selected from pancreatic cancer, ovarian cancer, lung cancer, endometrial cancer and mesothelioma.
  • the mesothelin-positive cancer is a dual-positive cancer.
  • a method of treating an individual having a mesothelin-positive cancer comprising administering to the individual an effective amount of an immunoconjugate as in any of the above embodiments.
  • the mesothelin-positive cancer is selected from pancreatic cancer, ovarian cancer, lung cancer, endometrial cancer, and mesothelioma.
  • the mesothelin-positive cancer is a dual-positive cancer.
  • the method further comprises administering an additional therapeutic agent to the individual.
  • the additional therapeutic agent is gemcitabine.
  • the additional therapeutic agent is an anti-MUC16 antibody conjugated to a cytotoxic agent.
  • a method of inhibiting proliferation of a mesothelin-positive cell comprising exposing the cell to an immunoconjugate as in any of the above embodiments under conditions permissive for binding of the immunoconjugate to mesothelin on the surface of the cell, thereby inhibiting proliferation of the cell.
  • the cell is a pancreatic, ovarian, lung, mesothelioma, or endometrial cell.
  • the cell is a dual-positive cell.
  • the invention provides an antibody as in any of the above
  • the antibody is conjugated to a label.
  • the label is a positron emitter.
  • the positron emitter is 89 Zr.
  • a method of detecting human mesothelin in a biological sample comprising contacting the biological sample with an anti-mesothelin antibody as in any of the above embodiments under conditions permissive for binding of the anti-mesothelin antibody to a naturally occurring human mesothelin, and detecting whether a complex is formed between the anti-mesothelin antibody and a naturally occurring human mesothelin in the biological sample.
  • the anti-mesothelin antibody comprises (a) HVR-Hl, HVR-H2, HVR-H3, HVR-Ll, HVR-L2 and HVR-L3 of the antibody produced by hybridoma 19C3 having ATCC Accession No.
  • the biological sample is a pancreatic cancer sample, ovarian cancer sample, lung cancer sample, endometrial cancer sample, or mesothelioma sample.
  • the method comprises performing immunohistochemistry on a tissue section.
  • the biological sample is serum.
  • a method for detecting a mesothelin-positive cancer comprising administering a labeled anti-mesothelin antibody, wherein the anti- mesothelin antibody is as in any of the above embodiments, to a subject having or suspected of having a mesothelin-positive cancer, and detecting the labeled anti-mesothelin antibody in the subject, wherein detection of the labeled anti-mesothelin antibody indicates a mesothelin- positive cancer in the subject.
  • the labeled anti-mesothelin antibody comprises an anti-mesothelin antibody conjugated to a positron emitter.
  • the positron emitter is 89 Zr.
  • Figure 1 shows that mesothelin is generated by proteolytic cleavage of a precursor protein into a 31 kDa shed component (referred to as megakaryocyte potentiating factor, or MPF) and a 40 kDa mesothelin component.
  • MPF megakaryocyte potentiating factor
  • the latter component may remain associated with the cell surface but may also be shed.
  • "CHO" represent the four glycosylation sites, one in MPF and three in mesothelin.
  • Figure 2 shows a graphic representation of the levels of human mesothelin gene expression in various tissues, as described in Example A.
  • Figure 3 shows properties of anti-mesothelin monoclonal antibodies isolated as described in Example B.
  • Figure 4 shows an alignment of the variable light chain region sequences of murine antibody 7D9 (mu7D9) and humanized variants thereof (7D9.vl and 7D9.v3).
  • Figure 5 shows an alignment of the variable heavy chain region sequences of murine antibody 7D9 (mu7D9) and humanized variants thereof (7D9.vl and 7D9.v3).
  • Figure 6 shows properties of chimeric and humanized variants of 7D9, as described in Example C.
  • Figure 7 shows an alignment of the variable light chain region sequences of murine antibody 22A10 (22A10) and humanized variants thereof (hu22A10graft and 22A10.v83).
  • Figure 8 shows an alignment of the variable heavy chain region sequences of murine antibody 22A10 (22A10) and humanized variants thereof (hu22A10graft and 22A10.v83).
  • Figure 9A shows Scatchard analysis of humanized variants of 22A10 on stably mesothelin- transfected BJAB cells, as described in Example C.
  • Figure 9B shows immunoprecipitation of mesothelin by humanized variants of 22A10 from the same stably transfected BJAB cells, as described in Example C.
  • Figure 10A shows the sequences of hypervariable and framework regions of humanized variants of 7D9.
  • Figure 10B shows the sequences of hypervariable and framework regions of humanized variants of 22A10.
  • Figure 11 shows sequence homology among mesothelin from different species, as described in Example D.
  • Figure 11 discloses SEQ ID NOS 43 and 46-48, respectively, in order of appearance.
  • Figure 12 shows cross-reactivities of h7D9.v3 and h22A10.v83 with mesothelin from different species, as described in Example D.
  • Figure 13 shows the affinities of humanized anti-mesothelin antibodies as determined by Scatchard analysis of transfected cell lines stably expressing mesothelin and cell lines expressing endogenous mesothelin, as described in Example E.
  • Figure 14 shows the results of competition assays between antibody 7D9 or 22A10 and the other monoclonal antibodies listed in Figure 3, as described in Example F.
  • Figure 15 shows chimeric mesothelin constructs used for epitope mapping (drawn to scale), as described in Example G.
  • Figure 15 discloses "EVEK,” “DAEQ,” and “DVER” as SEQ ID NOS 51-53, respectively.
  • Figure 16 shows the results of FACS to assess binding of 7D9 and 22A10 to cells expressing chimeric mesothelin, as described in Example G.
  • Figure 17 shows a mutational strategy for identifying the amino acids to which h7D9.v3 and h22A10.v83 bind, as described in Example G.
  • Figure 17 discloses "EVEK” as SEQ ID NO: 51; "Humanl32-212,” “Cyno 132-212,” “Ratl32-212,” and “Mouse 132-212” as SEQ ID NOS 54-57, respectively; human and mouse “MUT1,” “MUT3,” “MUT6,” “MUT7,” “MUT9,” “MUT10,” “MUT13,” and “MUT15,” as SEQ ID NOS 58-73, respectively; and “STKD” and "SVKD” as SEQ ID NOS 73 and 74, respectively.
  • Figure 18A shows the results of FACS to assess binding of h7D9.v3 and h22A10.v83 to cells expressing human mesothelin mutants, as described in Example G.
  • Figure 18B shows the results of FACS to assess binding of h7D9.v3 to cells expressing cynomolgus monkey mesothelin mutants, as described in Example G.
  • Figure 19 shows the key amino acid residues within the epitopes to which
  • Figure 20 shows binding of h7D9.v3 to glycosylated mesothelin, as described in Example H.
  • Figure 21 shows the results of two assays to determine whether antibodies 19C3, 7D9 and 22A10 block binding of mesothelin to MUC16 and vice versa, as described in Example I.
  • Figure 22 shows expression of mesothelin in pancreatic ductal adenocarcinoma by immunohistochemistry (IHC), as described in Example J.
  • Figure 23 shows expression of mesothelin in ovarian serous adenocarcinoma tumors by immunohistochemistry (IHC), as described in Example J.
  • Figure 24 shows expression of mesothelin in non-small cell lung cancer (NSCLC) adenocarcinoma by immunohistochemistry (IHC), as described in Example J.
  • NSCLC non-small cell lung cancer
  • IHC immunohistochemistry
  • Figure 25 shows expression of mesothelin in tissues from cynomolgus monkey (right panels) by immunohistochemistry (IHC), as described in Example J.
  • Figure 26 shows that the immunoconjugate h7D9.v3-vcMMAE demonstrates efficacy in HP AC pancreatic xenografts, as described in Example L.
  • Figure 27 shows that the immunoconjugate h7D9.v3-vcMMAE demonstrates efficacy in a primary pancreatic xenograft, as described in Example M.
  • Figure 28 shows that the immunoconjugate h7D9.v3-vcMMAE demonstrates efficacy in an ovarian tumor xenograft model, as described in Example N.
  • Figure 29 shows that the immunoconjugate h7D9.v3-vcMMAE demonstrates efficacy in a lung squamous cell carcinoma xenograft model, as described in Example O.
  • Figure 30 shows that the efficacy of the immunoconjugate h7D9.v3-vcMMAE against human mesothelin is similar to that of the immunoconjugate h22A10.v83-vcMMAE against cynomolgus monkey mesothelin in transfected BJAB xenograft tumor models, as described in Example P.
  • Figure 31 shows that the efficacy of the immunoconjugate h7D9.v3-vcMMAE is similar to that of the immunoconjugate h22A10.v83-vcMMAE in mesothelioma and ovarian tumor models, as described in Example P.
  • Figure 32 shows that MUC16 forms a complex with mesothelin, and the two proteins are co-shed from dual-positive cell lines, as described in Example Q.
  • Figure 33 shows that 19C3, but not 7D9, displaces pre-bound MUC16 from
  • acceptor human framework for the purposes herein is a framework comprising the amino acid sequence of a light chain variable domain (VL) framework or a heavy chain variable domain (VH) framework derived from a human immunoglobulin framework or a human consensus framework, as defined below.
  • An acceptor human framework "derived from” a human immunoglobulin framework or a human consensus framework may comprise the same amino acid sequence thereof, or it may contain amino acid sequence changes. In some embodiments, the number of amino acid changes are 10 or less, 9 or less, 8 or less, 7 or less, 6 or less, 5 or less, 4 or less, 3 or less, or 2 or less.
  • the VL acceptor human framework is identical in sequence to the VL human immunoglobulin framework sequence or human consensus framework sequence.
  • Binding affinity refers to the strength of the sum total of noncovalent interactions between a single binding site of a molecule (e.g., an antibody) and its binding partner (e.g., an antigen). Unless indicated otherwise, as used herein, "binding affinity” refers to intrinsic binding affinity which reflects a 1 : 1 interaction between members of a binding pair (e.g., antibody and antigen).
  • the affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (Kd). Affinity can be measured by common methods known in the art, including those described herein. Specific illustrative and exemplary embodiments for measuring binding affinity are described in the following.
  • an “affinity matured” antibody refers to an antibody with one or more alterations in one or more hypervariable regions (HVRs), compared to a parent antibody which does not possess such alterations, such alterations resulting in an improvement in the affinity of the antibody for antigen.
  • HVRs hypervariable regions
  • anti-mesothelin antibody and "an antibody that binds to mesothelin” refer to an antibody that is capable of binding mesothelin with sufficient affinity such that the antibody is useful as a diagnostic and/or therapeutic agent in targeting mesothelin.
  • the extent of binding of an anti-mesothelin antibody to an unrelated, non- mesothelin protein is less than about 10% of the binding of the antibody to mesothelin as measured, e.g., by a radioimmunoassay (RIA).
  • RIA radioimmunoassay
  • an antibody that binds to mesothelin has a dissociation constant (Kd) of ⁇ ⁇ , ⁇ 100 nM, ⁇ 10 nM, ⁇ 1 nM, ⁇ 0.1 nM, ⁇ 0.01 nM, or ⁇ 0.001 nM (e.g. 10 ⁇ 8 M or less, e.g. from 10 ⁇ 8 M to 10 "13 M, e.g., from 10 "9
  • an anti-mesothelin antibody binds to an epitope of mesothelin that is conserved among mesothelin from different species.
  • antibody is used herein in the broadest sense and encompasses various antibody structures, including but not limited to monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), and antibody fragments so long as they exhibit the desired antigen-binding activity.
  • antibody fragment refers to a molecule other than an intact antibody that comprises a portion of an intact antibody and that binds the antigen to which the intact antibody binds.
  • antibody fragments include but are not limited to Fv, Fab, Fab',
  • Fab'-SH, F(ab') 2 diabodies; linear antibodies; single-chain antibody molecules (e.g. scFv); and multispecific antibodies formed from antibody fragments.
  • an "antibody that binds to the same epitope” as a reference antibody refers to an antibody that blocks binding of the reference antibody to its antigen in a competition assay by 50% or more, and conversely, the reference antibody blocks binding of the antibody to its antigen in a competition assay by 50% or more.
  • An exemplary competition assay is provided herein.
  • cancer and “cancerous” refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth/proliferation.
  • examples of cancer include, but are not limited to, carcinoma, lymphoma (e.g., Hodgkin's and non-
  • chimeric antibody refers to an antibody in which a portion of the heavy and/or light chain is derived from a particular source or species, while the remainder of the heavy and/or light chain is derived from a different source
  • the "class" of an antibody refers to the type of constant domain or constant region possessed by its heavy chain.
  • the heavy chain constant domains that correspond to the different classes of immunoglobulins are called ⁇ , ⁇ , ⁇ , ⁇ , and ⁇ , respectively.
  • cytotoxic agent refers to a substance that inhibits or prevents a cellular function and/or causes cell death or destruction. Cytotoxic agents include, but are not
  • radioactive isotopes e.g., At , 1 , 1 , Y , Re , Re , Sm , Bi , P ,
  • chemotherapeutic agents or drugs e.g., methotrexate, adriamicin, vinca alkaloids (vincristine, vinblastine, etoposide), doxorubicin, melphalan, mitomycin C, chlorambucil, daunorubicin or other intercalating agents
  • growth inhibitory agents e.g., methotrexate, adriamicin, vinca alkaloids (vincristine, vinblastine, etoposide), doxorubicin, melphalan, mitomycin C, chlorambucil, daunorubicin or other intercalating agents
  • growth inhibitory agents e.g., enzymes and fragments thereof such as nucleolytic enzymes; antibiotics; toxins such as small molecule toxins or enzymatically active toxins of bacterial, fungal, plant or animal origin, including fragments and/or variants thereof; and the various antitumor or anticancer agents disclosed below.
  • dual-positive cancer refers to a cancer comprising cells that are both mesothelin- and MUC16-positive.
  • the term "dual-positive cell” refers to a cell that expresses both mesothelin and
  • Antibody effector functions refer to those biological activities attributable to the Fc region of an antibody, which vary with the antibody isotype. Examples of antibody effector functions include: Clq binding and complement dependent cytotoxicity (CDC); Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; down regulation of cell surface receptors (e.g. B cell receptor); and B cell activation.
  • an "effective amount" of an agent refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result.
  • epitopope refers to the particular site on an antigen molecule to which an antibody binds.
  • Fc region herein is used to define a C-terminal region of an immunoglobulin heavy chain that contains at least a portion of the constant region.
  • the term includes native sequence Fc regions and variant Fc regions.
  • a human IgG heavy chain Fc region extends from Cys226, or from Pro230, to the carboxyl-terminus of the heavy chain.
  • the C-terminal lysine (Lys447) of the Fc region may or may not be present.
  • EU numbering system also called the EU index, as described in Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD, 1991.
  • FR Framework or "FR” refers to variable domain residues other than hypervariable region (HVR) residues.
  • the FR of a variable domain generally consists of four FR domains: FR1, FR2, FR3, and FR4. Accordingly, the HVR and FR sequences generally appear in the following sequence in VH (or VL): FR1-H1(L1)-FR2-H2(L2)-FR3-H3(L3)-FR4.
  • full length antibody “intact antibody,” and “whole antibody” are used herein interchangeably to refer to an antibody having a structure substantially similar to a native antibody structure or having heavy chains that contain an Fc region as defined herein.
  • glycosylated forms of mesothelin refers to naturally occurring forms of mesothelin that are post-translationally modified by the addition of carbohydrate residues.
  • host cell refers to cells into which exogenous nucleic acid has been introduced, including the progeny of such cells.
  • Host cells include “transformants” and “transformed cells,” which include the primary transformed cell and progeny derived therefrom without regard to the number of passages. Progeny may not be completely identical in nucleic acid content to a parent cell, but may contain mutations. Mutant progeny that have the same function or biological activity as screened or selected for in the originally transformed cell are included herein.
  • a "human antibody” is one which possesses an amino acid sequence which corresponds to that of an antibody produced by a human or a human cell or derived from a non-human source that utilizes human antibody repertoires or other human antibody-encoding sequences. This definition of a human antibody specifically excludes a humanized antibody comprising non-human antigen-binding residues.
  • a "human consensus framework” is a framework which represents the most commonly occurring amino acid residues in a selection of human immunoglobulin VL or VH framework sequences.
  • the selection of human immunoglobulin VL or VH sequences is from a subgroup of variable domain sequences.
  • the subgroup of sequences is a subgroup as in Kabat et al., Sequences of Proteins of Immunological Interest, Fifth Edition, NIH
  • the subgroup is subgroup kappa I as in Kabat et al, supra. In one embodiment, for the VH, the subgroup is subgroup III as in Kabat et al, supra.
  • a “humanized” antibody refers to a chimeric antibody comprising amino acid residues from non-human HVRs and amino acid residues from human FRs.
  • a humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the HVRs (e.g., CDRs) correspond to those of a non-human antibody, and all or substantially all of the FRs correspond to those of a human antibody.
  • a humanized antibody optionally may comprise at least a portion of an antibody constant region derived from a human antibody.
  • a "humanized form" of an antibody, e.g., a non-human antibody refers to an antibody that has undergone humanization.
  • hypervariable region refers to each of the regions of an antibody variable domain which are hypervariable in sequence and/or form structurally defined loops ("hypervariable loops").
  • native four-chain antibodies comprise six HVRs; three in the VH (HI, H2, H3), and three in the VL (LI, L2, L3).
  • HVRs generally comprise amino acid residues from the hypervariable loops and/or from the "complementarity determining regions" (CDRs), the latter being of highest sequence variability and/or involved in antigen recognition.
  • CDRs complementarity determining regions
  • Exemplary hypervariable loops occur at amino acid residues 26-32 (LI), 50-52 (L2), 91-96 (L3), 26-32 (HI), 53-55 (H2), and 96-101 (H3).
  • Exemplary CDRs CDR-L1, CDR-L2, CDR-L3, CDR-H1, CDR-H2, and CDR-H3 occur at amino acid residues 24-34 of LI, 50-56 of L2, 89-97 of L3, 31-35B of HI, 50-65 of H2, and 95-102 of H3.
  • CDRs generally comprise the amino acid residues that form the hypervariable loops.
  • CDRs also comprise "specificity determining residues,” or "SDRs,” which are residues that contact antigen. SDRs are contained within regions of the CDRs called abbreviated-CDRs, or a-CDRs.
  • Exemplary a-CDRs (a-CDR-Ll, a- CDR-L2, a-CDR-L3, a-CDR-Hl, a-CDR-H2, and a-CDR-H3) occur at amino acid residues 31- 34 of LI, 50-55 of L2, 89-96 of L3, 31-35B of HI, 50-58 of H2, and 95-102 of H3.
  • HVR residues and other residues in the variable domain are numbered herein according to Kabat et al., supra.
  • An "immunoconjugate" is an antibody conjugated to one or more heterologous molecule(s), including but not limited to a cytotoxic agent.
  • mammals include, but are not limited to, domesticated animals (e.g., cows, sheep, cats, dogs, and horses), primates (e.g., humans and non-human primates such as monkeys), rabbits, and rodents (e.g., mice and rats).
  • domesticated animals e.g., cows, sheep, cats, dogs, and horses
  • primates e.g., humans and non-human primates such as monkeys
  • rabbits e.g., mice and rats
  • rodents e.g., mice and rats.
  • the individual or subject is a human.
  • an “isolated antibody” is one which has been separated from a component of its natural environment.
  • an antibody is purified to greater than 95% or 99% purity as determined by, for example, electrophoretic (e.g., SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis) or chromatographic (e.g., ion exchange or reverse phase HPLC).
  • electrophoretic e.g., SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis
  • chromatographic e.g., ion exchange or reverse phase HPLC
  • isolated nucleic acid refers to a nucleic acid molecule that has been separated from a component of its natural environment.
  • An isolated nucleic acid includes a nucleic acid molecule contained in cells that ordinarily contain the nucleic acid molecule, but the nucleic acid molecule is present extrachromosomally or at a chromosomal location that is different from its natural chromosomal location.
  • isolated nucleic acid encoding an anti-mesothelin antibody refers to one or more nucleic acid molecules encoding antibody heavy and light chains (or fragments thereof), including such nucleic acid molecule(s) in a single vector or separate vectors, and such nucleic acid molecule(s) present at one or more locations in a host cell.
  • mesothelin refers to any native, mature mesothelin which results from processing of a mesothelin precursor protein in a cell.
  • the term includes mesothelin from any vertebrate source, including mammals such as primates (e.g. humans and cynomolgus monkeys) and rodents (e.g., mice and rats), unless otherwise indicated.
  • the term also includes naturally occurring variants of mesothelin, e.g., splice variants or allelic variants.
  • SEQ ID NO:42 SEQ ID NO:42
  • an exemplary human mesothelin is shown in SEQ ID NO:43. Further exemplary mesothelin sequences are described herein.
  • mesothelin-positive cancer refers to a cancer comprising cells that express mesothelin on their surface.
  • the term “mesothelin-positive cell” refers to a cell that expresses mesothelin on its surface.
  • the term "monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical and/or bind the same epitope, except for possible variant antibodies, e.g., containing naturally occurring mutations or arising during production of a monoclonal antibody preparation, such variants generally being present in minor amounts.
  • polyclonal antibody preparations typically include different antibodies directed against different determinants (epitopes)
  • each monoclonal antibody of a monoclonal antibody preparation is directed against a single determinant on an antigen.
  • the modifier "monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies to be used in accordance with the present invention may be made by a variety of techniques, including but not limited to the hybridoma method, recombinant DNA methods, phage-display methods, and methods utilizing transgenic animals containing all or part of the human immunoglobulin loci, such methods and other exemplary methods for making monoclonal antibodies being described herein.
  • MUC16-positive cancer refers to a cancer comprising cells that express
  • MUC16-positive cell refers to a cell that expresses MUC16 on its surface.
  • a “naked antibody” refers to an antibody that is not conjugated to a heterologous moiety (e.g., a cytotoxic moiety) or radiolabel. The naked antibody may be present in a pharmaceutical formulation.
  • Native antibodies refer to naturally occurring immunoglobulin molecules with varying structures.
  • native IgG antibodies are heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light chains and two identical heavy chains that are disulfide-bonded. From N- to C-terminus, each heavy chain has a variable region
  • VH variable heavy domain
  • CHI heavy chain variable domain
  • CL constant light domain
  • variable light domain
  • lambda
  • package insert is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, combination therapy, contraindications and/or warnings concerning the use of such therapeutic products.
  • Percent (%) amino acid sequence identity with respect to a reference polypeptide sequence is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in the reference polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared.
  • % amino acid sequence identity values are generated using the sequence comparison computer program ALIGN-2.
  • the ALIGN-2 sequence comparison computer program was authored by Genentech, Inc., and the source code has been filed with user documentation in the U.S. Copyright Office, Washington D.C., 20559, where it is registered under U.S. Copyright Registration No. TXU510087.
  • the ALIGN-2 program is publicly available from Genentech, Inc., South San Francisco, California, or may be compiled from the source code.
  • the ALIGN-2 program should be compiled for use on a UNLX operating system, including digital UNIX V4.0D. All sequence comparison parameters are set by the ALIGN-2 program and do not vary.
  • % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B is calculated as follows:
  • pharmaceutical formulation refers to a preparation which is in such form as to permit the biological activity of an active ingredient contained therein to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered.
  • a “pharmaceutically acceptable carrier” refers to an ingredient in a pharmaceutical formulation, other than an active ingredient, which is nontoxic to a subject.
  • pharmaceutically acceptable carrier includes, but is not limited to, a buffer, excipient, stabilizer, or preservative.
  • treatment refers to clinical intervention in an attempt to alter the natural course of the individual being treated, and can be performed either for prophylaxis or during the course of clinical pathology. Desirable effects of treatment include, but are not limited to, preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastasis, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis.
  • antibodies of the invention are used to delay development of a disease or to slow the progression of a disease.
  • variable region refers to the domain of an antibody heavy or light chain that is involved in binding the antibody to antigen.
  • the variable domains of the heavy chain and light chain (VH and VL, respectively) of a native antibody generally have similar structures, with each domain comprising four conserved framework regions (FRs) and three hypervariable regions (HVRs).
  • FRs conserved framework regions
  • HVRs hypervariable regions
  • antibodies that bind a particular antigen may be isolated using a VH or VL domain from an antibody that binds the antigen to screen a library of complementary VL or VH domains, respectively. See, e.g., Portolano et al., J.
  • vector refers to a nucleic acid molecule capable of propagating another nucleic acid to which it is linked.
  • the term includes the vector as a self- replicating nucleic acid structure as well as the vector incorporated into the genome of a host cell into which it has been introduced.
  • Certain vectors are capable of directing the expression of nucleic acids to which they are operatively linked. Such vectors are referred to herein as "expression vectors.”
  • the invention is based, in part, on antibodies that bind to mesothelin and immunoconjugates comprising such antibodies.
  • Antibodies and immunoconjugates of the invention are useful, e.g., for the diagnosis or treatment of mesothelin-positive cancers.
  • the invention provides isolated antibodies that bind to mesothelin.
  • Naturally occurring mesothelin results from cleavage of a mesothelin precurson protein in a cell, generating mesothelin and megakaryocyte potentiating factor (MPF), as shown in Figure 1.
  • Mesothelin contains a C-terminal truncation relative to the precursor protein. Such truncation may allow for attachment of a GPI anchor.
  • Mesothelin may remain associated with the cell surface, e.g., via the GPI anchor, or mesothelin may be released from the cell (e.g., the GPI anchor may be cleaved by an as yet unidentified enzyme) to produce shed mesothelin in cell culture or animal serum.
  • SEQ ID NO:42 An exemplary naturally occurring human mesothelin precursor protein sequence is provided in SEQ ID NO:42, and the corresponding mesothelin sequence is shown in SEQ ID NO:43 (corresponding to amino acids 296-580 of SEQ ID NO:42).
  • An alternative mesothelin sequence corresponds to amino acids 296-598 of SEQ ID NO:42.
  • SEQ ID NO:44 is a naturally occurring variant of SEQ ID NO:42, the processing of which results in a mesothelin having the sequence of SEQ ID NO:45.
  • SEQ ID NO:45 contains an eight amino acid insertion at amino acid 116 relative to SEQ ID NO:43.
  • the variant form of mesothelin shown in SEQ ID NO:45 appears to comprise -5% of mesothelin transcripts in tumor cell lines.
  • an anti-mesothelin antibody has at least one or more of the following characteristics, in any combination:
  • an antibody that does not block binding of mesothelin to MUC16 is an antibody that enhances binding of mesothelin to MUC16.
  • an anti-mesothelin antibody binds to an epitope of SEQ ID NO:43 comprising E153 and D174.
  • the anti-mesothelin antibody further has one or more of the following characteristics, in any combination:
  • (c) binds mesothelin with an affinity of ⁇ 5 nM, or alternatively ⁇ 1 nM, or alternatively ⁇ 0.5 nM, and optionally > 0.0001 nM.
  • an antibody that does not block binding of mesothelin to MUC16 enhances binding of mesothelin to MUC16 and/or the antibody binds with an affinity of ⁇ 1 nM.
  • An exemplary antibody having the above characteristics is 7D9 and humanized variants thereof, such as h7D9.v3, disclosed herein.
  • the mesothelin to which an anti-mesothelin antibody binds is human mesothelin.
  • an anti-mesothelin antibody binds to an epitope of SEQ ID NO:43 comprising E211.
  • the anti-mesothelin antibody further has one or more of the following characteristics:
  • (b) binds mesothelin with an affinity of ⁇ 5 nM, or alternatively ⁇ 1 nM, or alternatively ⁇ 0.5 nM, and optionally > 0.0001 nM.
  • an antibody that does not block binding of mesothelin to MUC16 enhances binding of mesothelin to MUC16, and/or the antibody binds with an affinity of ⁇ 1 nM.
  • An exemplary antibody having the above characteristics is 22A10 and humanized variants thereof, such as 22A10.v83, disclosed herein.
  • the mesothelin to which an anti-mesothelin antibody binds is human mesothelin, cynomolgus monkey mesothelin, and/or rat mesothelin.
  • an anti-mesothelin antibody in another embodiment, an anti-mesothelin antibody:
  • (b) binds mesothelin with an affinity of ⁇ 5 nM, or alternatively ⁇ 1 nM, or alternatively ⁇ 0.5 nM, or alternatively ⁇ 0.1 nM, and optionally > 0.0001 nM.
  • the antibody blocks binding of mesothelin to MUC16 and/or binds to an epitope within amino acids 1-64 or 1-70 of SEQ ID NO:43. In one such embodiment, the antibody displaces MUC16 bound to mesothelin.
  • An exemplary antibody having the above characteristics is 19C3, disclosed herein.
  • the mesothelin to which an anti-mesothelin antibody binds is human mesothelin.
  • an anti-mesothelin antibody “binds to an epitope of SEQ ID NO:43 comprising E153 and D174,” or “binds to an epitope of SEQ ID NO:43 comprising E211,” those residues are mutated in a polypeptide comprising SEQ ID NO:43, and binding of the antibody to the mutated polypeptide expressed in 293 cells is tested by FACS as described in Example G, wherein a substantial reduction (> 70% reduction) or elimination of binding of the antibody to the mutated polypeptide indicates that the antibody binds to an epitope of SEQ ID NO:43 comprising E153 and D174, or comprising E211.
  • tagged human mesothelin is expressed in CHO cells, purified (by way of the tag) and further separated according to charge on a Mono S column into fractions with high (fraction Al 1), medium (A12), low (Bl) and low-to-none (B5)
  • a MUC 16 binding assay is performed, as follows. Specifically, a biotinylated fragment of MUC 16 (encompassing three of the mucin repeats) is incubated with A431 cells stably expressing mesothelin in the presence or absence of anti-mesothelin antibody, and the level of MUC16-biotin binding to the cells is determined by FACS with streptavidin-PE. The MUC 16 binding site of mesothelin has been tentatively mapped to the first 64 amino acids of mesothelin (Kaneko et al., J. Biol Chem. 284:3739-49 (2009)).
  • PC3 cells stably expressing MUC 16 are incubated with purified mesothelin-his8 ("his8" disclosed as SEQ ID NO: 49) preincubated with anti-mesothelin antibodies, and binding of purified mesothelin-his8: antibody complexes to the MUC16-expressing cells is detected by FACS using an Alexa-647 conjugated anti-His6 antibody ("His6" disclosed as SEQ ID NO: 50). If in either of the above assays, the FACS signal is >50% lower in the presence of anti-mesothelin antibody than in the absence, then that antibody is considered to block binding of mesothelin to MUC 16.
  • the FACS signal is not decreased by >50% in the presence of anti-mesothelin antibody, then that antibody is considered to not block binding of mesothelin to MUC 16. If in the latter of the above assays, the FACS signal is increased in the presence of anti-mesothelin antibody than in the absence, then that antibody is considered to enhance binding of mesothelin to MUC 16.
  • an anti-mesothelin antibody "binds with an affinity of ⁇ 5 nM, or alternatively ⁇ 1 nM, or alternatively ⁇ 0.5 nM, or alternatively ⁇ 0.1 nM" affinity is determined according to a Biacore assay as described herein in Section II. A.1. Specifically, Kd is measured using surface plasmon resonance assays using a BIACORE -2000 or a BIACORE -3000 (BIAcore, Inc., Piscataway, NJ) at 25°C with immobilized antigen CM5 chips at -10 response units (RU).
  • a Biacore assay as described herein in Section II. A.1. Specifically, Kd is measured using surface plasmon resonance assays using a BIACORE -2000 or a BIACORE -3000 (BIAcore, Inc., Piscataway, NJ) at 25°C with immobilized antigen CM5 chips at -10 response units (RU).
  • carboxymethylated dextran biosensor chips (CM5, BIACORE, Inc.) are activated with N-ethyl-N - (3-dimethylaminopropyl)-carbodiimide hydrochloride (EDC) and N- hydroxysuccinimide (NHS) according to the supplier's instructions.
  • EDC N-ethyl-N - (3-dimethylaminopropyl)-carbodiimide hydrochloride
  • NHS N- hydroxysuccinimide
  • Fab form in PBS, pH 7.2, in the presence of increasing concentrations of antigen as measured in a spectrometer, such as a stop-flow equipped spectrophometer (Aviv Instruments) or a 8000- series SLM-AMINCOTM spectrophotometer (ThermoSpectronic) with a stirred cuvette.
  • a spectrometer such as a stop-flow equipped spectrophometer (Aviv Instruments) or a 8000- series SLM-AMINCOTM spectrophotometer (ThermoSpectronic) with a stirred cuvette.
  • a spectrometer such as a stop-flow equipped spectrophometer (Aviv Instruments) or a 8000- series SLM-AMINCOTM spectrophotometer (ThermoSpectronic) with a stirred cuvette.
  • the invention provides an anti-mesothelin antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO:20; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO:21; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:22; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 17; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 18; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 19.
  • HVR-H1 comprising the amino acid sequence of SEQ ID NO:20
  • HVR-H2 comprising the amino acid sequence of SEQ ID NO:21
  • HVR-H3 comprising the amino acid sequence of SEQ ID NO:22
  • HVR-L1 comprising the amino acid sequence of SEQ ID NO: 17
  • HVR-L2 comprising
  • the invention provides an antibody comprising at least one, at least two, or all three VH HVR sequences selected from (a) HVR-Hl comprising the amino acid sequence of SEQ ID NO:20; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO:21; and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:22.
  • the antibody comprises HVR-H3 comprising the amino acid sequence of SEQ ID NO:22.
  • the antibody comprises HVR-H3 comprising the amino acid sequence of SEQ ID NO:22 and HVR-L3 comprising the amino acid sequence of SEQ ID
  • the antibody comprises HVR-H3 comprising the amino acid sequence of SEQ ID NO:22, HVR-L3 comprising the amino acid sequence of SEQ ID NO: 19, and HVR-H2 comprising the amino acid sequence of SEQ ID NO:21.
  • HVR-H3 comprising the amino acid sequence of SEQ ID NO:22
  • HVR-L3 comprising the amino acid sequence of SEQ ID NO: 19
  • HVR-H2 comprising the amino acid sequence of SEQ ID NO:21.
  • the antibody comprises (a) HVR-Hl comprising the amino acid sequence of SEQ ID NO :20; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO :21 ; and (c) HVR- H3 comprising the amino acid sequence of SEQ ID NO:22.
  • the invention provides an antibody comprising at least one, at least two, or all three VL HVR sequences selected from (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 17; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 18; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 19.
  • the antibody comprises (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 17; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 18; and (c) HVR- L3 comprising the amino acid sequence of SEQ ID NO: 19.
  • an antibody of the invention comprises (a) a VH domain comprising at least one, at least two, or all three VH HVR sequences selected from (i) HVR-Hl comprising the amino acid sequence of SEQ ID NO:20, (ii) HVR-H2 comprising the amino acid sequence of SEQ ID NO:21, and (iii) HVR-H3 comprising an amino acid sequence selected from SEQ ID NO:22; and (b) a VL domain comprising at least one, at least two, or all three VL HVR sequences selected from (i) HVR-Ll comprising the amino acid sequence of SEQ ID NO: 17, (ii) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 18, and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 19.
  • the invention provides an antibody comprising (a) HVR-Hl comprising the amino acid sequence of SEQ ID NO:20; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO:21; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:22; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 17; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 18; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 19.
  • an anti-mesothelin antibody is humanized.
  • an anti-mesothelin antibody comprises HVRs as in any of the above
  • a human acceptor framework e.g. a human
  • the human acceptor framework is the human VL kappa I consensus (VL KI ) framework and/or the VH framework VH AT A, which differs from the human VH subgroup III consensus (VH m ) at 3 positions: R71A, N73T, and L78A (Carter et al, Proc. Natl. Acad. Sci. USA 89:4285 (1992)).
  • an anti-mesothelin antibody comprises HVRs as in any of the above embodiments, and further comprises a light chain variable domain comprising a framework FR2 sequence of SEQ ID NO:25 and an FR3 sequence of SEQ ID NO:27.
  • the light chain variable domain framework is a modified human VL kappa I consensus (VL KI ) framework having FR2 sequence of SEQ ID NO:25 and an FR3 sequence of SEQ ID NO:27.
  • an anti-mesothelin antibody comprises a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%) sequence identity to the amino acid sequence of SEQ ID NO:8.
  • VH heavy chain variable domain
  • a VH sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-mesothelin antibody comprising that sequence retains the ability to bind to mesothelin.
  • the anti-mesothelin antibody comprises the VH sequence of SEQ ID NO: 8, including post- translational modifications of that sequence.
  • the VH comprises one, two or three HVRs selected from: (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO:20, (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO:21, and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:22.
  • an anti-mesothelin antibody comprising a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO:4.
  • VL light chain variable domain
  • a VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%), or 99% identity contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-mesothelin antibody comprising that sequence retains the ability to bind to mesothelin.
  • the anti-mesothelin antibody comprises the VL sequence of SEQ ID NO:4, including post-translational modifications of that sequence.
  • the VL comprises one, two or three HVRs selected from (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 17; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO : 18 ; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO : 19.
  • an anti-mesothelin antibody comprising a VH as in any of the embodiments provided above, and a VL as in any of the embodiments provided above.
  • the antibody comprises the VH and VL sequences in SEQ ID NO: 8 and SEQ ID NO:4, respectively, including post-translational modifications of those sequences.
  • the invention provides an antibody that binds to the same epitope as an anti-mesothelin antibody provided herein.
  • an antibody is provided that binds to the same epitope as an anti-mesothelin antibody comprising a VH sequence of SEQ ID NO:8 and a VL sequence of SEQ ID NO:4.
  • an antibody that binds to an epitope of SEQ ID NO:43 from, within, or overlapping amino acids 152-175. In certain embodiments, an antibody is provided that binds to an epitope of SEQ ID NO:43 comprising E153 and D174. In certain such
  • the antibody binds to amino acid residues El 53 and D174.
  • an anti-mesothelin antibody is a monoclonal antibody, including a chimeric, humanized or human antibody.
  • an anti-mesothelin antibody is an antibody fragment, e.g., a Fv, Fab, Fab', scFv, diabody, or F(ab') 2 fragment.
  • the antibody is a substantially full length antibody, e.g., an IgGl antibody or other antibody class or isotype as defined herein.
  • an anti-mesothelin antibody may incorporate any of the features, singly or in combination, as described in Sections 1-7 below: Antibody 22A10 and other embodiments
  • the invention provides an anti-mesothelin antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-Hl comprising the amino acid sequence of SEQ ID NO:36; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO:37; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:38 or 39; (d) HVR- Ll comprising the amino acid sequence of SEQ ID NO: 33; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO:34; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO:35.
  • HVR-Hl comprising the amino acid sequence of SEQ ID NO:36
  • HVR-H2 comprising the amino acid sequence of SEQ ID NO:37
  • HVR-H3 comprising the amino acid sequence of SEQ ID NO:38 or 39
  • HVR- Ll comprising the amino acid sequence of SEQ ID NO: 33
  • the invention provides an antibody comprising at least one, at least two, or all three VH HVR sequences selected from (a) HVR-Hl comprising the amino acid sequence of SEQ ID NO:36; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO:37; and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:38 or 39.
  • the antibody comprises HVR-H3 comprising the amino acid sequence of SEQ ID NO:38 or 39.
  • the antibody comprises HVR-H3 comprising the amino acid sequence of SEQ ID NO:38 or 39, and HVR-L3 comprising the amino acid sequence of SEQ ID NO:35.
  • the antibody comprises HVR-H3 comprising the amino acid sequence of SEQ ID NO:38 or 39, HVR-L3 comprising the amino acid sequence of SEQ ID NO:35, and HVR-H2 comprising the amino acid sequence of SEQ ID NO:37.
  • the antibody comprises (a) HVR-Hl comprising the amino acid sequence of SEQ ID NO:36; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO:37; and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:38 or 39.
  • the invention provides an antibody comprising at least one, at least two, or all three VL HVR sequences selected from (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO:33; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO:34; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO:35.
  • the antibody comprises (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO:33; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO:34; and (c) HVR- L3 comprising the amino acid sequence of SEQ ID NO: 35.
  • an antibody of the invention comprises (a) a VH domain comprising at least one, at least two, or all three VH HVR sequences selected from (i) HVR-Hl comprising the amino acid sequence of SEQ ID NO:36, (ii) HVR-H2 comprising the amino acid sequence of SEQ ID NO:37, and (iii) HVR-H3 comprising the amino acid sequence of SEQ ID NO:38 or 39; and (b) a VL domain comprising at least one, at least two, or all three VL HVR sequences selected from (i) HVR-L1 comprising the amino acid sequence of SEQ ID NO:33, (ii) HVR-L2 comprising the amino acid sequence of SEQ ID NO:34, and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO:35.
  • the invention provides an antibody comprising (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO:36; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO:37; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:38 or 39; (d) HVR-Ll comprising the amino acid sequence of SEQ ID NO:33; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO:34; and (f) HVR-L3 comprising an amino acid sequence selected from SEQ ID NO:35.
  • an anti-mesothelin antibody is humanized.
  • an anti-mesothelin antibody comprises HVRs as in any of the above
  • an acceptor human framework e.g. a human
  • the human acceptor framework is VLKI and/or VH m acceptor framework.
  • an anti-mesothelin antibody comprises a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 16.
  • VH heavy chain variable domain
  • a VH sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%o, or 99% identity contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-mesothelin antibody comprising that sequence retains the ability to bind to mesothelin.
  • a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 16.
  • substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs).
  • the anti-mesothelin antibody comprises the VH sequence of SEQ ID NO: 16, including post-translational modifications of that sequence.
  • the VH comprises one, two or three HVRs selected from: (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO:36, (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO:37, and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:38 or 39.
  • an anti-mesothelin antibody comprising a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 12.
  • VL light chain variable domain
  • a VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%), 96%o, 97%), 98%o, or 99% identity contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-mesothelin antibody comprising that sequence retains the ability to bind to mesothelin.
  • the anti-mesothelin antibody comprises the VL sequence of SEQ ID NO: 12, including post-translational modifications of that sequence.
  • the VL comprises one, two or three HVRs selected from (a) HVR-Ll comprising the amino acid sequence of SEQ ID NO:33; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO:34; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO:35.
  • an anti-mesothelin antibody comprising a VH as in any of the embodiments provided above, and a VL as in any of the embodiments provided above.
  • the antibody comprises the VH and VL sequences in SEQ ID NO: 16 and SEQ ID NO: 12, respectively, including post-translational modifications of those sequences.
  • the invention provides an antibody that binds to the same epitope as an anti-mesothelin antibody provided herein.
  • an antibody is provided that binds to the same epitope as an anti-mesothelin antibody comprising a VH sequence of SEQ ID NO: 16 and a VL sequence of SEQ ID NO: 12.
  • an antibody that binds to an epitope of SEQ ID NO:43 from, within, or overlapping amino acids 211-327. In certain embodiments, an antibody is provided that binds to an epitope of SEQ ID NO:43 comprising E211. In certain such embodiments, the antibody binds to amino acid residue E211.
  • an anti-mesothelin antibody is a monoclonal antibody, including a chimeric, humanized or human antibody.
  • an anti-mesothelin antibody is an antibody fragment, e.g., a Fv, Fab, Fab', scFv, diabody, or F(ab') 2 fragment.
  • the antibody is a substantially full length antibody, e.g., an IgG2a antibody or other antibody class or isotype as defined herein.
  • an anti-mesothelin antibody may incorporate any of the features, singly or in combination, as described in Sections 1-7 below:
  • the invention provides an anti-mesothelin antibody comprising at least one, two, three, four, five, or six HVRs of the antibody produced by hybridoma 19C3 having ATCC Accession No. PTA-11464.
  • HVRs are delineated by the amino acid ranges corresponding to CDRs, as defined herein.
  • the invention provides an antibody comprising at least one, at least two, or all three VH HVR sequences of the antibody produced by hybridoma 19C3 having ATCC Accession No. PTA-11464.
  • the antibody comprises HVR-H3 of the antibody produced by hybridoma 19C3 having ATCC Accession No. PTA-11464.
  • the antibody comprises HVR-H3 and HVR-L3 of the antibody produced by hybridoma 19C3 having ATCC Accession No. PTA-11464.
  • the antibody comprises HVR-H3, HVR-L3, and HVR-H2 of the antibody produced by hybridoma 19C3 having ATCC Accession No. PTA-11464.
  • the antibody comprises HVR-H1, HVR-H2, and HVR-H3 of the antibody produced by hybridoma 19C3 having ATCC Accession No. PTA-11464.
  • the invention provides an antibody comprising at least one, at least two, or all three VL HVR sequences of the antibody produced by hybridoma 19C3 having ATCC Accession No. PTA-11464.
  • the antibody comprises HVR-L1,
  • HVR-L2 and HVR-L3 of the antibody produced by hybridoma 19C3 having ATCC Accession
  • an antibody of the invention comprises (a) a VH domain comprising at least one, at least two, or all three VH HVR sequences of the antibody produced by hybridoma 19C3 having ATCC Accession No. PTA-11464; and (b) a VL domain comprising at least one, at least two, or all three VL HVR sequences of the antibody produced by hybridoma 19C3 having ATCC Accession No. PTA-11464.
  • the invention provides an antibody comprising HVR-H1, HVR-H2, HVR-H3, HVR-L1, HVR-L2, and HVR-L3 of the antibody produced by hybridoma 19C3 having ATCC Accession No. PTA-11464.
  • an anti-mesothelin antibody is humanized.
  • the antibody is a humanized form of the antibody produced by hybridoma 19C3 having ATCC Accession No. PTA-11464.
  • an anti-mesothelin antibody comprises HVRs as in any of the above embodiments, and further comprises an acceptor human framework, e.g. a human immunoglobulin framework or a human consensus framework.
  • an anti-mesothelin antibody comprises a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%) sequence identity to the VH of the antibody produced by hybridoma 19C3 having ATCC Accession No. PTA-11464.
  • VH sequence contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-mesothelin antibody comprising that sequence retains the ability to bind to mesothelin.
  • the anti-mesothelin antibody comprises the VH sequence of the antibody produced by hybridoma 19C3 having ATCC Accession No. PTA- 11464, including post-translational modifications of that sequence.
  • the VH comprises one, two, or three HVRs selected from HVR-H1, HVR-H2, and HVR-H3 of the antibody produced by hybridoma 19C3 having ATCC Accession No. PTA- 11464.
  • an anti-mesothelin antibody comprising a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the VL of the antibody produced by hybridoma 19C3 having ATCC Accession No. PTA-11464.
  • VL light chain variable domain
  • a VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-mesothelin antibody comprising that sequence retains the ability to bind to mesothelin.
  • a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in the VL of the antibody produced by hybridoma 19C3 having ATCC Accession No. PTA-11464.
  • the substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs).
  • the anti-mesothelin antibody comprises the VL sequence of the antibody produced by hybridoma 19C3 having ATCC Accession No. PTA-11464, including post-translational modifications of that sequence.
  • the VL comprises one, two or three HVRs selected from HVR-L1, HVR-L2, and HVR-L3 of the antibody produced by hybridoma 19C3 having ATCC Accession No. PTA-11464.
  • an anti-mesothelin antibody comprising a VH as in any of the embodiments provided above, and a VL as in any of the embodiments provided above.
  • the antibody comprises the VH and VL sequences of the antibody produced by hybridoma 19C3 having ATCC Accession No. PTA- 11464, respectively, including post-translational modifications of those sequences.
  • the invention provides an antibody that binds to the same epitope as an anti-mesothelin antibody provided herein.
  • an antibody is provided that binds to the same epitope as the antibody produced by hybridoma 19C3 having ATCC Accession No. PTA-11464.
  • an anti-mesothelin antibody is a monoclonal antibody, including a chimeric, humanized or human antibody.
  • an anti-mesothelin antibody is an antibody fragment, e.g., a Fv, Fab, Fab', scFv, diabody, or F(ab') 2 fragment.
  • the antibody is a substantially full length antibody, e.g., an IgG2b antibody or other antibody class or isotype as defined herein.
  • an anti-mesothelin antibody may incorporate any of the features, singly or in combination, as described in Sections 1-7 below:
  • an antibody provided herein has a dissociation constant (Kd) of ⁇ ⁇ , ⁇ 100 nM, ⁇ 10 nM, ⁇ 1 nM, ⁇ 0.1 nM, ⁇ 0.01 nM, or ⁇ 0.001 nM, and optionally is > 10 ⁇ 13 M. (e.g. 10 ⁇ 8 M or less, e.g. from 10 ⁇ 8 M to 10 ⁇ 13 M, e.g., from 10 "9 M to 10 "13 M).
  • Kd dissociation constant
  • Kd is measured by a radiolabeled antigen binding assay (RIA) performed with the Fab version of an antibody of interest and its antigen as described by the following assay.
  • Solution binding affinity of Fabs for antigen is measured by equilibrating Fab with a minimal concentration of ( 125 I)-labeled antigen in the presence of a titration series of unlabeled antigen, then capturing bound antigen with an anti-Fab antibody-coated plate (see, e.g., Chen et al, J. Mol. Biol. 293:865-881(1999)).
  • MICROTITER multi-well plates (Thermo Scientific) are coated overnight with 5 ⁇ g/ml of a capturing anti-Fab antibody (Cappel Labs) in 50 mM sodium carbonate (pH 9.6), and subsequently blocked with 2% (w/v) bovine serum albumin in PBS for two to five hours at room temperature (approximately 23°C).
  • a capturing anti-Fab antibody Cappel Labs
  • bovine serum albumin in PBS for two to five hours at room temperature (approximately 23°C).
  • a non-adsorbent plate (Nunc #269620), 100 pM or
  • 26 pM [ 125 I]-antigen are mixed with serial dilutions of a Fab of interest (e.g., consistent with assessment of the anti-VEGF antibody, Fab-12, in Presta et al, Cancer Res. 57:4593-4599
  • the Fab of interest is then incubated overnight; however, the incubation may continue for a longer period (e.g., about 65 hours) to ensure that equilibrium is reached. Thereafter, the mixtures are transferred to the capture plate for incubation at room temperature (e.g., for one hour). The solution is then removed and the plate washed eight times with 0.1% polysorbate 20 (TWEEN-20 ® ) in PBS. When the plates have dried, 150 ⁇ /well of scintillant
  • Kd is measured using surface plasmon resonance assays using a BIACORE ® -2000 or a BIACORE ® -3000 (BIAcore, Inc., Piscataway, NJ) at 25°C with immobilized antigen CM5 chips at -10 response units (RU).
  • carboxymethylated dextran biosensor chips (CM5, BIACORE, Inc.) are activated with N-ethyl- N'- (3-dimethylaminopropyl)-carbodiimide hydrochloride (EDC) and N-hydroxysuccinimide (NHS) according to the supplier's instructions.
  • EDC N-ethyl- N'- (3-dimethylaminopropyl)-carbodiimide hydrochloride
  • NHS N-hydroxysuccinimide
  • Antigen is diluted with 10 mM sodium acetate, pH 4.8, to 5 ⁇ g/ml (-0.2 ⁇ ) before injection at a flow rate of 5 ⁇ /minute to achieve approximately 10 response units (RU) of coupled protein. Following the injection of antigen, 1 M ethanolamine is injected to block unreacted groups.
  • association rates (k on ) and dissociation rates (k 0 ff) are calculated using a simple one-to-one Langmuir binding model (BIACORE Evaluation Software version 3.2) by simultaneously fitting the association and dissociation sensorgrams.
  • the equilibrium dissociation constant (Kd) is calculated as the ratio k 0 ff/k on See, e.g., Chen et al, J. Mol. Biol. 293:865-881
  • an antibody provided herein is an antibody fragment.
  • Antibody fragments include, but are not limited to, Fab, Fab', Fab'-SH, F(ab') 2 , Fv, and scFv fragments, and other fragments described below.
  • Fab fragment antigen binding fragment
  • Fab' fragment antigen binding fragment
  • Fab'-SH fragment antigen binding fragment
  • Fv fragment antigen binding fragment
  • scFv fragments fragment antigen binding fragments
  • U.S. Patent Nos. 5,571,894 and 5,587,458 For discussion of Fab and F(ab') 2 fragments comprising salvage receptor binding epitope residues and having increased in vivo half- life, see U.S.
  • Diabodies are antibody fragments with two antigen-binding sites that may be bivalent or bispecific. See, for example, EP 404,097; WO 1993/01161; Hudson et al, Nat. Med. 9: 129- 134 (2003); and Hollinger et al, Proc. Natl. Acad. Sci. USA 90: 6444-6448 (1993). Triabodies and tetrabodies are also described in Hudson et al, Nat. Med. 9: 129-134 (2003).
  • Single-domain antibodies are antibody fragments comprising all or a portion of the heavy chain variable domain or all or a portion of the light chain variable domain of an antibody.
  • a single-domain antibody is a human single-domain antibody (Domantis, Inc., Waltham, MA; see, e.g., U.S. Patent No. 6,248,516 Bl).
  • Antibody fragments can be made by various techniques, including but not limited to proteolytic digestion of an intact antibody as well as production by recombinant host cells (e.g. E. coli or phage), as described herein.
  • recombinant host cells e.g. E. coli or phage
  • an antibody provided herein is a chimeric antibody.
  • Certain chimeric antibodies are described, e.g., in U.S. Patent No. 4,816,567; and Morrison et al, Proc. Natl. Acad. Sci. USA, 81 :6851-6855 (1984)).
  • a chimeric antibody comprises a non-human variable region (e.g., a variable region derived from a mouse, rat, hamster, rabbit, or non-human primate, such as a monkey) and a human constant region.
  • a chimeric antibody is a "class switched" antibody in which the class or subclass has been changed from that of the parent antibody. Chimeric antibodies include antigen-binding fragments thereof.
  • a chimeric antibody is a humanized antibody.
  • a non-human antibody is humanized to reduce immunogenicity to humans, while retaining the specificity and affinity of the parental non-human antibody.
  • a humanized antibody comprises one or more variable domains in which HVRs, e.g., CDRs, (or portions thereof) are derived from a non-human antibody, and FRs (or portions thereof) are derived from human antibody sequences.
  • HVRs e.g., CDRs, (or portions thereof) are derived from a non-human antibody
  • FRs or portions thereof
  • a humanized antibody optionally will also comprise at least a portion of a human constant region.
  • some FR residues in a humanized antibody are substituted with corresponding residues from a non-human antibody (e.g., the antibody from which the HVR residues are derived), e.g., to restore or improve antibody specificity or affinity.
  • a non-human antibody e.g., the antibody from which the HVR residues are derived
  • Human framework regions that may be used for humanization include but are not limited to: framework regions selected using the "best-fit” method (see, e.g., Sims et al. J. Immunol. 151 :2296 (1993)); framework regions derived from the consensus sequence of human antibodies of a particular subgroup of light or heavy chain variable regions (see, e.g., Carter et al. Proc. Natl. Acad. Sci. USA, 89:4285 (1992); and Presta et al. J. Immunol,
  • an antibody provided herein is a human antibody.
  • Human antibodies can be produced using various techniques known in the art. Human antibodies are described generally in van Dijk and van de Winkel, Curr. Opin. Pharmacol. 5: 368-74 (2001) and Lonberg, Curr. Opin. Immunol. 20:450-459 (2008).
  • Human antibodies may be prepared by administering an immunogen to a transgenic animal that has been modified to produce intact human antibodies or intact antibodies with human variable regions in response to antigenic challenge. Such animals typically contain all or a portion of the human immunoglobulin loci, which replace the endogenous
  • immunoglobulin loci or which are present extrachromosomally or integrated randomly into the animal's chromosomes. In such transgenic mice, the endogenous immunoglobulin loci have generally been inactivated.
  • endogenous immunoglobulin loci have generally been inactivated.
  • Human antibodies can also be made by hybridoma-based methods. Human myeloma and mouse-human heteromyeloma cell lines for the production of human monoclonal antibodies have been described. (See, e.g., Kozbor J. Immunol, 133 : 3001 (1984); Brodeur et al., Monoclonal Antibody Production Techniques and Applications, pp. 51 -63 (Marcel Dekker, Inc., New York, 1987); and Boerner et al, J. Immunol., 147: 86 (1991).) Human antibodies generated via human B-ceil hybridoma technology are also described in Li et al, Proc, Natl, Acad. Sci. USA, 103:3557-3562 (2006).
  • Additional methods include those described, for example, in U.S. Patent No. 7, 189,826 (describing production of monoclonal human IgM antibodies from hybridoma cell lines) and Ni, Xiandai Mianyixue, 26(4):265-268 (2006) (describing human-human hybridomas).
  • Human hybridoma technology Trioma technology
  • Vollmers and Brandlein, Histology and Histopathology, 20(3):927-937 (2005) and Vollmers and Brandlein, Methods and Findings in Experimental and Clinical Pharmacology, 27(3): 185-91 (2005).
  • Human antibodies may also be generated by isolating Fv clone variable domain sequences selected from human-derived phage display libraries. Such variable domain sequences may then be combined with a desired human constant domain. Techniques for selecting human antibodies from antibody libraries are described below.
  • Antibodies of the invention may be isolated by screening combinatorial libraries for antibodies with the desired activity or activities. For example, a variety of methods are known in the art for generating phage display libraries and screening such libraries for antibodies possessing the desired binding characteristics. Such methods are reviewed, e.g., in
  • repertoires of VH and VL genes are separately cloned by polymerase chain reaction (PCR) and recombined randomly in phage libraries, which can then be screened for antigen-binding phage as described in Winter et al, Ann. Rev. Immunol, 12: 433-455 (1994).
  • Phage typically display antibody fragments, either as single-chain Fv (scFv) fragments or as Fab fragments.
  • naive repertoire can be cloned (e.g., from human) to provide a single source of antibodies to a wide range of non-self and also self antigens without any immunization as described by Griffiths et al., EMBO J, 12: 725-734 (1993).
  • naive libraries can also be made synthetically by cloning unrearranged V-gene segments from stem cells, and using PCR primers containing random sequence to encode the highly variable CDR3 regions and to accomplish rearrangement in vitro, as described by Hoogenboom and Winter, J. Mol. Biol, 227: 381-388 (1992).
  • Patent publications describing human antibody phage libraries include, for example: US Patent No. 5,750,373, and US Patent Publication Nos. 2005/0079574, 2005/0119455, 2005/0266000, 2007/0117126, 2007/0160598, 2007/0237764, 2007/0292936, and 2009/0002360.
  • Antibodies or antibody fragments isolated from human antibody libraries are considered human antibodies or human antibody fragments herein.
  • an antibody provided herein is a multispecific antibody, e.g. a bispecific antibody.
  • Multispecific antibodies are monoclonal antibodies that have binding specificities for at least two different sites. In certain embodiments, one of the binding specificities is for mesothelin and the other is for any other antigen.
  • bispecific antibodies may bind to two different epitopes of mesothelin. Bispecific antibodies may also be used to localize cytotoxic agents to cells which express mesothelin. Bispecific antibodies can be prepared as full length antibodies or antibody fragments.
  • Multispecific antibodies include, but are not limited to, recombinant co-expression of two immunoglobulin heavy chain-light chain pairs having different specificities (see Milstein and Cuello, Nature 305: 537 (1983)), WO 93/08829, and Traunecker et al, EMBO J. 10: 3655 (1991)), and "knob-in-hole” engineering (see, e.g., U.S. Patent No. 5,731,168).
  • Multi-specific antibodies may also be made by engineering electrostatic steering effects for making antibody Fc-heterodimeric molecules (WO 2009/089004A1); cross- linking two or more antibodies or fragments (see, e.g., US Patent No.
  • the antibody or fragment herein also includes a "Dual Acting FAb” or “DAF” comprising an antigen binding site that binds to mesothelin as well as another, different antigen (see, US 2008/0069820, for example).
  • amino acid sequence variants of the antibodies provided herein are contemplated. For example, it may be desirable to improve the binding affinity and/or other biological properties of the antibody.
  • Amino acid sequence variants of an antibody may be prepared by introducing appropriate modifications into the nucleotide sequence encoding the antibody, or by peptide synthesis. Such modifications include, for example, deletions from, and/or insertions into and/or substitutions of residues within the amino acid sequences of the antibody. Any combination of deletion, insertion, and substitution can be made to arrive at the final construct, provided that the final construct possesses the desired characteristics, e.g., antigen-binding.
  • antibody variants having one or more amino acid substitutions are provided.
  • Sites of interest for substitutional mutagenesis include the HVRs and FRs.
  • amino acid side chain classes Amino acid substitutions may be introduced into an antibody of interest and the products screened for a desired activity, e.g., retained/improved antigen binding, decreased
  • Amino acids may be grouped according to common side-chain properties:
  • Non-conservative substitutions will entail exchanging a member of one of these classes for another class.
  • substitutional variant involves substituting one or more hypervariable region residues of a parent antibody (e.g. a humanized or human antibody).
  • a parent antibody e.g. a humanized or human antibody
  • the resulting variant(s) selected for further study will have modifications (e.g., improvements) in certain biological properties (e.g., increased affinity, reduced immunogenicity) relative to the parent antibody and/or will have substantially retained certain biological properties of the parent antibody.
  • An exemplary substitutional variant is an affinity matured antibody, which may be conveniently generated, e.g., using phage display-based affinity maturation techniques such as those described herein. Briefly, one or more HVR residues are mutated and the variant antibodies displayed on phage and screened for a particular biological activity (e.g. binding affinity).
  • Alterations may be made in HVRs, e.g., to improve antibody affinity. Such alterations may be made in HVR "hotspots," i.e., residues encoded by codons that undergo mutation at high frequency during the somatic maturation process (see, e.g., Chowdhury, Methods Mol. Biol. 207: 179-196 (2008)), and/or SDRs (a-CDRs), with the resulting variant VH or VL being tested for binding affinity.
  • HVR "hotspots” i.e., residues encoded by codons that undergo mutation at high frequency during the somatic maturation process (see, e.g., Chowdhury, Methods Mol. Biol. 207: 179-196 (2008)
  • SDRs a-CDRs
  • affinity maturation diversity is introduced into the variable genes chosen for maturation by any of a variety of methods (e.g., error-prone PCR, chain shuffling, or oligonucleotide-directed mutagenesis).
  • a secondary library is then created. The library is then screened to identify any antibody variants with the desired affinity.
  • HVR-directed approaches in which several HVR residues (e.g., 4-6 residues at a time) are randomized.
  • HVR residues involved in antigen binding may be specifically identified, e.g., using alanine scanning mutagenesis or modeling.
  • CDR-H3 and CDR-L3 in particular are often targeted.
  • substitutions, insertions, or deletions may occur within one or more HVRs so long as such alterations do not substantially reduce the ability of the antibody to bind antigen.
  • conservative alterations e.g., conservative substitutions as provided herein
  • Such alterations may be outside of HVR "hotspots" or SDRs.
  • each HVR either is unaltered, or contains no more than one, two or three amino acid substitutions.
  • a useful method for identification of residues or regions of an antibody that may be targeted for mutagenesis is called "alanine scanning mutagenesis" as described by Cunningham and Wells (1989) Science, 244: 1081-1085.
  • a residue or group of target residues e.g., charged residues such as arg, asp, his, lys, and glu
  • a neutral or negatively charged amino acid e.g., alanine or polyalanine
  • a crystal structure of an antigen-antibody complex is used to identify contact points between the antibody and antigen. Such contact residues and neighboring residues may be targeted or eliminated as candidates for substitution. Variants may be screened to determine whether they contain the desired properties.
  • Amino acid sequence insertions include amino- and/or carboxyl-terminal fusions ranging in length from one residue to polypeptides containing a hundred or more residues, as well as intrasequence insertions of single or multiple amino acid residues.
  • terminal insertions include an antibody with an N-terminal methionyl residue.
  • Other insertional variants of the antibody molecule include the fusion to the N- or C -terminus of the antibody to an enzyme (e.g. for ADEPT) or a polypeptide which increases the serum half-life of the antibody.
  • an antibody provided herein is altered to increase or decrease the extent to which the antibody is glycosylated.
  • Addition or deletion of glycosylation sites to an antibody may be conveniently accomplished by altering the amino acid sequence such that one or more glycosylation sites is created or removed.
  • the carbohydrate attached thereto may be altered.
  • Native antibodies produced by mammalian cells typically comprise a branched, biantennary oligosaccharide that is generally attached by an N-linkage to Asn297 of the CH2 domain of the Fc region. See, e.g., Wright et al. TIBTECH 15:26-32 (1997).
  • oligosaccharide may include various carbohydrates, e.g., mannose, N-acetyl glucosamine (GlcNAc), galactose, and sialic acid, as well as a fucose attached to a GlcNAc in the "stem" of the biantennary oligosaccharide structure.
  • modifications of the oligosaccharide in an antibody of the invention may be made in order to create antibody variants with certain improved properties.
  • antibody variants having a carbohydrate structure that lacks fucose attached (directly or indirectly) to an Fc region.
  • the amount of fucose in such antibody may be from 1% to 80%, from 1% to 65%, from 5% to 65%> or from 20% to 40%.
  • the amount of fucose is determined by calculating the average amount of fucose within the sugar chain at Asn297, relative to the sum of all glycostructures attached to Asn 297 (e. g. complex, hybrid and high mannose structures) as measured by MALDI-TOF mass spectrometry, as described in WO 2008/077546, for example.
  • Asn297 refers to the asparagine residue located at about position 297 in the Fc region (Eu numbering of Fc region residues); however, Asn297 may also be located about ⁇ 3 amino acids upstream or downstream of position 297, i.e., between positions 294 and 300, due to minor sequence variations in antibodies.
  • Such fucosylation variants may have improved ADCC function. See, e.g., US Patent Publication Nos. US 2003/0157108 (Presta, L.); US 2004/0093621 (Kyowa Hakko Kogyo Co., Ltd). Examples of publications related to "defucosylated” or "fucose-deficient" antibody variants include: US 2003/0157108; WO 2000/61739; WO 2001/29246; US
  • Examples of cell lines capable of producing defucosylated antibodies include Lecl3 CHO cells deficient in protein fucosylation (Ripka et al. Arch. Biochem. Biophys. 249:533-545 (1986); US Pat Appl No US 2003/0157108 Al, Presta, L; and WO 2004/056312 Al, Adams et al, especially at Example 11), and knockout cell lines, such as alpha- 1,6-fucosyltransferase gene, FUT8, knockout CHO cells (see, e.g., Yamane-Ohnuki et al. Biotech. Bioeng. 87: 614 (2004); Kanda, Y. et al, Biotechnol. Bioeng., 94(4):680-688 (2006); and WO2003/085107).
  • Antibodies variants are further provided with bisected oligosaccharides, e.g., in which a biantennary oligosaccharide attached to the Fc region of the antibody is bisected by GlcNAc. Such antibody variants may have reduced fucosylation and/or improved ADCC function. Examples of such antibody variants are described, e.g., in WO 2003/011878 (Jean-Mairet et al); US Patent No. 6,602,684 (Umana et al); and US 2005/0123546 (Umana et al). Antibody variants with at least one galactose residue in the oligosaccharide attached to the Fc region are also provided. Such antibody variants may have improved CDC function. Such antibody variants are described, e.g., in WO 1997/30087 (Patel et al); WO 1998/58964 (Raju, S.); and WO 1999/22764 (Raju, S.).
  • one or more amino acid modifications may be introduced into the Fc region of an antibody provided herein, thereby generating an Fc region variant.
  • the Fc region variant may comprise a human Fc region sequence ⁇ e.g., a human IgGl, IgG2, IgG3 or IgG4 Fc region) comprising an amino acid modification (e.g. a substitution) at one or more amino acid positions.
  • the invention contemplates an antibody variant that possesses some but not all effector functions, which make it a desirable candidate for applications in which the half life of the antibody in vivo is important yet certain effector functions (such as complement and ADCC) are unnecessary or deleterious.
  • In vitro and/or in vivo cytotoxicity assays can be conducted to confirm the reduction/depletion of CDC and/or ADCC activities.
  • Fc receptor (FcR) binding assays can be conducted to ensure that the antibody lacks FcyR binding (hence likely lacking ADCC activity), but retains FcRn binding ability.
  • NK cells express FcyRIII only, whereas monocytes express FcyRI, FcyRII and FcyRIII.
  • FcR expression on hematopoietic cells is summarized in Table 3 on page 464 of Ravetch and Kinet, Annu. Rev. Immunol. 9:457-492 (1991).
  • Non- limiting examples of in vitro assays to assess ADCC activity of a molecule of interest is described in U.S. Patent No. 5,500,362 (see, e.g. Hellstrom, I. et al. Proc. Nat 7 Acad. Sci. USA 83:7059-7063 (1986)) and Hellstrom, I et al, Proc.
  • non-radioactive assays methods may be employed (see, for example, ACTITM non-radioactive cytotoxicity assay for flow cytometry (CellTechnology, Inc. Mountain View, CA; and CytoTox 96 ® non-radioactive cytotoxicity assay (Promega, Madison, WI).
  • Useful effector cells for such assays include peripheral blood mononuclear cells (PBMC) and Natural Killer (NK) cells.
  • ADCC activity of the molecule of interest may be assessed in vivo, e.g., in a animal model such as that disclosed in Clynes et al. Proc. Nat 'l Acad. Sci. USA 95:652-656 (1998).
  • Clq binding assays may also be carried out to confirm that the antibody is unable to bind Clq and hence lacks CDC activity. See, e.g., Clq and C3c binding ELISA in WO 2006/029879 and WO 2005/100402.
  • a CDC assay may be performed (see, for example, Gazzano-Santoro et al., J. Immunol.
  • FcRn binding and in vivo clearance/half life determinations can also be performed using methods known in the art (see, e.g., Petkova, S.B. et al, Int'l. Immunol. 18(12): 1759-1769 (2006)).
  • Antibodies with reduced effector function include those with substitution of one or more of Fc region residues 238, 265, 269, 270, 297, 327 and 329 (U.S. Patent No. 6,737,056).
  • Fc mutants include Fc mutants with substitutions at two or more of amino acid positions 265, 269, 270, 297 and 327, including the so-called "DANA" Fc mutant with substitution of residues 265 and 297 to alanine (US Patent No. 7,332,581).
  • an antibody variant comprises an Fc region with one or more amino acid substitutions which improve ADCC, e.g., substitutions at positions 298, 333, and/or 334 of the Fc region (EU numbering of residues).
  • alterations are made in the Fc region that result in altered (i.e., either improved or diminished) Clq binding and/or Complement Dependent Cytotoxicity (CDC), e.g., as described in US Patent No. 6,194,551, WO 99/51642, and Idusogie et al. J. Immunol. 164: 4178-4184 (2000).
  • CDC Complement Dependent Cytotoxicity
  • FcRn neonatal Fc receptor
  • Those antibodies comprise an Fc region with one or more substitutions therein which improve binding of the Fc region to FcRn.
  • Fc variants include those with substitutions at one or more of Fc region residues: 238, 256, 265, 272, 286, 303, 305, 307, 311, 312, 317, 340, 356, 360, 362, 376, 378, 380, 382, 413, 424 or 434, e.g., substitution of Fc region residue 434 (US Patent No. 7,371 ,826).
  • cysteine engineered antibodies e.g., "thioMAbs”
  • one or more residues of an antibody are substituted with cysteine residues.
  • the substituted residues occur at accessible sites of the antibody.
  • reactive thiol groups are thereby positioned at accessible sites of the antibody and may be used to conjugate the antibody to other moieties, such as drug moieties or linker-drug moieties, to create an immunoconjugate, as described further herein.
  • any one or more of the following residues may be substituted with cysteine: V205 (Kabat numbering) of the light chain; Al 18 (EU numbering) of the heavy chain; and S400 (EU numbering) of the heavy chain Fc region.
  • Cysteine engineered antibodies may be generated as described, e.g., in U.S. Patent No.
  • an antibody provided herein may be further modified to contain additional nonproteinaceous moieties that are known in the art and readily available.
  • the moieties suitable for derivatization of the antibody include but are not limited to water soluble polymers.
  • water soluble polymers include, but are not limited to, polyethylene glycol (PEG), copolymers of ethylene glycol/propylene glycol, carboxymethylcellulose, dextran, polyvinyl alcohol, polyvinyl pyrrolidone, poly-1, 3- dioxolane, poly-1, 3, 6-trioxane, ethylene/maleic anhydride copolymer, polyaminoacids (either homopolymers or random copolymers), and dextran or poly(n- vinyl pyrrolidone)poly ethylene glycol, propropylene glycol homopolymers, prolypropylene oxide/ethylene oxide co-polymers, polyoxyethylated polyols (e.g., glycerol),
  • PEG poly
  • Polyethylene glycol propionaldehyde may have advantages in manufacturing due to its stability in water.
  • the polymer may be of any molecular weight, and may be branched or unbranched.
  • the number of polymers attached to the antibody may vary, and if more than one polymer are attached, they can be the same or different molecules. In general, the number and/or type of polymers used for derivatization can be determined based on considerations including, but not limited to, the particular properties or functions of the antibody to be improved, whether the antibody derivative will be used in a therapy under defined conditions, etc.
  • conjugates of an antibody and nonproteinaceous moiety that may be selectively heated by exposure to radiation are provided.
  • the nonproteinaceous moiety is a carbon nanotube (Kam et al., Proc. Natl. Acad. Sci. USA 102: 11600-11605 (2005)).
  • the radiation may be of any wavelength, and includes, but is not limited to, wavelengths that do not harm ordinary cells, but which heat the nonproteinaceous moiety to a temperature at which cells proximal to the antibody-nonproteinaceous moiety are killed.
  • Antibodies may be produced using recombinant methods and compositions, e.g., as described in U.S. Patent No. 4,816,567.
  • isolated nucleic acid encoding an anti-mesothelin antibody described herein is provided.
  • Such nucleic acid may encode an amino acid sequence comprising the VL and/or an amino acid sequence comprising the VH of the antibody (e.g., the light and/or heavy chains of the antibody).
  • one or more vectors e.g., expression vectors
  • a host cell comprising such nucleic acid is provided.
  • a host cell comprises (e.g., has been transformed with): (1) a vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL of the antibody and an amino acid sequence comprising the VH of the antibody, or (2) a first vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL of the antibody and a second vector comprising a nucleic acid that encodes an amino acid sequence comprising the VH of the antibody.
  • the host cell is eukaryotic, e.g. a Chinese Hamster Ovary (CHO) cell or lymphoid cell (e.g., Y0, NSO, Sp20 cell).
  • a method of making an anti-mesothelin antibody comprises culturing a host cell comprising a nucleic acid encoding the antibody, as provided above, under conditions suitable for expression of the antibody, and optionally recovering the antibody from the host cell (or host cell culture medium).
  • nucleic acid encoding an antibody is isolated and inserted into one or more vectors for further cloning and/or expression in a host cell.
  • nucleic acid may be readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the antibody).
  • Suitable host cells for cloning or expression of antibody-encoding vectors include prokaryotic or eukaryotic cells described herein.
  • antibodies may be produced in bacteria, in particular when glycosylation and Fc effector function are not needed.
  • U.S. Patent Nos. 5,648,237, 5,789,199, and 5,840,523. See also Charlton, Methods in Molecular Biology, Vol. 248 (B.K.C. Lo, ed., Humana Press, Totowa, NJ, 2003), pp. 245-254, describing expression of antibody fragments in E. coli.
  • the antibody may be isolated from the bacterial cell paste in a soluble fraction and can be further purified.
  • eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for antibody-encoding vectors, including fungi and yeast strains whose glycosylation pathways have been "humanized,” resulting in the production of an antibody with a partially or fully human glycosylation pattern. See Gerngross, Nat. Biotech. 22: 1409-1414 (2004), and Li et al, Nat. Biotech. 24:210-215 (2006).
  • Suitable host cells for the expression of glycosylated antibody are also derived from multicellular organisms (invertebrates and vertebrates). Examples of invertebrate cells include plant and insect cells. Numerous baculoviral strains have been identified which may be used in conjunction with insect cells, particularly for transfection of Spodoptera frugiperda cells.
  • Plant cell cultures can also be utilized as hosts. See, e.g., US Patent Nos. 5,959,177, 6,040,498, 6,420,548, 7,125,978, and 6,417,429 (describing PLANTIBODIESTM technology for producing antibodies in transgenic plants).
  • Vertebrate cells may also be used as hosts.
  • mammalian cell lines that are adapted to grow in suspension may be useful.
  • Other examples of useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-7); human embryonic kidney line (293 or 293 cells as described, e.g., in Graham et al., J. Gen Virol. 36:59 (1977)); baby hamster kidney cells (BHK); mouse Sertoli cells (TM4 cells as described, e.g., in Mather, Biol. Reprod.
  • monkey kidney cells (CV1); African green monkey kidney cells (VERO-76); human cervical carcinoma cells (HELA); canine kidney cells (MDCK; buffalo rat liver cells (BRL 3 A); human lung cells (W138); human liver cells (Hep G2); mouse mammary tumor (MMT 060562); TRI cells, as described, e.g., in Mather et al, Annals N. Y. Acad. Sci. 383:44-68 (1982); MRC 5 cells; and FS4 cells.
  • Other useful mammalian host cell lines include Chinese hamster ovary (CHO) cells, including DHFR " CHO cells (Urlaub et al., Proc. Natl. Acad. Sci.
  • Anti-mesothelin antibodies provided herein may be identified, screened for, or characterized for their physical/chemical properties and/or biological activities by various assays known in the art.
  • an antibody of the invention is tested for its antigen binding activity, e.g., by known methods such as ELISA, FACS or Western blot.
  • competition assays may be used to identify an antibody that competes with any of the antibodies described herein for binding to mesothelin.
  • a competing antibody binds to the same epitope (e.g., a linear or a conformational epitope) that is bound by an antibody described herein.
  • epitope e.g., a linear or a conformational epitope
  • Detailed exemplary methods for mapping an epitope to which an antibody binds are provided in Morris (1996) "Epitope Mapping Protocols," in Methods in Molecular Biology vol. 66 (Humana Press, Totowa, NJ).
  • immobilized mesothelin is incubated in a solution comprising a first labeled antibody that binds to mesothelin (e.g., any of the antibodies described herein) and a second unlabeled antibody that is being tested for its ability to compete with the first antibody for binding to mesothelin.
  • the second antibody may be present in a hybridoma supernatant.
  • immobilized mesothelin is incubated in a solution comprising the first labeled antibody but not the second unlabeled antibody.
  • the invention also provides immunoconjugates comprising an anti-mesothelin antibody herein conjugated to one or more cytotoxic agents, such as chemotherapeutic agents or drugs, growth inhibitory agents, toxins (e.g., protein toxins, enzymatically active toxins of bacterial, fungal, plant, or animal origin, or fragments thereof), or radioactive isotopes.
  • cytotoxic agents such as chemotherapeutic agents or drugs, growth inhibitory agents, toxins (e.g., protein toxins, enzymatically active toxins of bacterial, fungal, plant, or animal origin, or fragments thereof), or radioactive isotopes.
  • an immunoconjugate is an antibody-drug conjugate (ADC) in which an antibody is conjugated to one or more drugs, including but not limited to a maytansinoid (see U.S. Patent Nos. 5,208,020, 5,416,064 and European Patent EP 0 425 235 Bl); an auristatin such as monomethylauristatin drug moieties DE and DF (MMAE and MMAF) (see U.S. Patent Nos. 5,635,483 and 5,780,588, and 7,498,298); a dolastatin; a calicheamicin or derivative thereof (see U.S. Patent Nos. 5,712,374, 5,714,586, 5,739,116, 5,767,285, 5,770,701, 5,770,710, 5,773,001, and 5,877,296; Hinman et al, Cancer Res.
  • ADC antibody-drug conjugate
  • an immunoconjugate comprises an antibody as described herein conjugated to an enzymatically active toxin or fragment thereof, including but not limited to diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, modeccin A chain, alpha- sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin, and the tricothecenes.
  • an enzymatically active toxin or fragment thereof including but not limited to diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain
  • an immunoconjugate comprises an antibody as described herein conjugated to a radioactive atom to form a radioconjugate.
  • a radioactive atom to form a radioconjugate.
  • isotopes are available for the production of radioconjugates. Examples include At , 1 , 1 , Y 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 , Pb 212 and radioactive isotopes of Lu.
  • radioconjugate is used for detection, it may comprise a radioactive atom for scintigraphic studies, for example tc99m or 1123, or a spin label for nuclear magnetic resonance (NMR) imaging (also known as magnetic resonance imaging, mri), such as iodine- 123 again, iodine- 131, indium- 111, fluorine- 19 , carbon- 13 , nitrogen- 15 , oxygen- 17 , gadolinium, manganese or iron.
  • NMR nuclear magnetic resonance
  • Conjugates of an antibody and cytotoxic agent may be made using a variety of bifunctional protein coupling agents such as N-succinimidyl-3-(2-pyridyldithio) propionate (SPDP), succinimidyl-4-(N-maleimidomethyl) cyclohexane-l-carboxylate (SMCC), iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HC1), active esters (such as disuccinimidyl suberate), aldehydes (such as glutaraldehyde), bis-azido compounds (such as bis (p-azidobenzoyl) hexanediamine), bis-diazonium derivatives (such as bis-(p-diazoniumbenzoyl)-ethylenediamine), diisocyanates (such as toluene 2,6-diisocyanate), and bis-active fluorine compounds (such
  • Carbon- 14-labeled l-isothiocyanatobenzyl-3-methyldiethylene triaminepentaacetic acid is an exemplary chelating agent for conjugation of radionucleotide to the antibody. See W094/11026.
  • the linker may be a "cleavable linker" facilitating release of a cytotoxic drug in the cell.
  • an acid-labile linker, peptidase-sensitive linker, photolabile linker, dimethyl linker or disulfide-containing linker (Chari et al., Cancer Res. 52: 127-131 (1992); U.S. Patent No. 5,208,020) may be used.
  • the immunuoconjugates or ADCs herein expressly contemplate, but are not limited to such conjugates prepared with cross-linker reagents including, but not limited to, BMPS, EMCS, GMBS, HBVS, LC-SMCC, MBS, MPBH, SBAP, SIA, SIAB, SMCC, SMPB, SMPH, sulfo-EMCS, sulfo-GMBS, sulfo-KMUS, sulfo-MBS, sulfo-SIAB, sulfo-SMCC, and sulfo- SMPB, and SVSB (succinimidyl-(4-vinylsulfone)benzoate) which are commercially available (e.g., from Pierce Biotechnology, Inc., Rockford, IL., U.S.A).
  • cross-linker reagents including, but not limited to, BMPS, EMCS, GMBS, HBVS,
  • an immunoconjugate comprises an antibody of the invention conjugated to dolastatin or a dolastatin peptidic analog or derivative, e.g., an auristatin (US Pat. Nos. 5635483; 5780588).
  • dolastatins and auristatins have been shown to interfere with microtubule dynamics, GTP hydrolysis, and nuclear and cellular division (Woyke et al (2001) Antimicrob. Agents and Chemother. 45(12):3580-3584) and have anticancer (US Pat. No. 5663149) and antifungal activity (Pettit et al (1998) Antimicrob. Agents Chemother. 42:2961- 2965).
  • the dolastatin or auristatin drug moiety may be attached to the antibody through the N (amino) terminus or the C (carboxyl) terminus of the peptidic drug moiety (WO 02/088172).
  • Exemplary auristatin embodiments include the N-terminus linked monomethylauristatin drug moieties DE and DF. (See US Patent Nos. 7,659,241, 7,498,298, and 7,745,394.)
  • a peptidic drug moiety may be selected from Formulas D E and D F below:
  • R is selected from H and Ci-Cg alkyl
  • R is selected from H, Ci-C 8 alkyl, C 3 -C 8 carbocycle, aryl, Ci-C 8 alkyl-aryl, Ci-C 8 alkyl-(C 3 -C 8 carbocycle), C 3 -C 8 heterocycle and Ci-C 8 alkyl-(C 3 -C 8 heterocycle);
  • R 4 is selected from H, Ci-C 8 alkyl, C 3 -C 8 carbocycle, aryl, Ci-C 8 alkyl-aryl, Ci-C 8 alkyl-(C 3 -C 8 carbocycle), C 3 -C 8 heterocycle and Ci-C 8 alkyl-(C 3 -C 8 heterocycle);
  • R 5 is selected from H and methyl; or R 4 and R 5 jointly form a carbocyclic ring and have the formula -(CR a R b ) n - wherein R a and R b are independently selected from H, Ci-C 8 alkyl and C 3 -C 8 carbocycle and n is selected from 2, 3, 4, 5 and 6;
  • R 6 is selected from H and Ci-C 8 alkyl
  • R 7 is selected from H, Ci-C 8 alkyl, C 3 -C 8 carbocycle, aryl, Ci-C 8 alkyl-aryl, Ci-C 8 alkyl-(C 3 -C 8 carbocycle), C 3 -C 8 heterocycle and Ci-C 8 alkyl-(C 3 -C 8 heterocycle);
  • each R is independently selected from H, OH, Ci-C 8 alkyl, C 3 -C 8 carbocycle and O- (Ci-Cg alkyl);
  • R 9 is selected from H and Ci-C 8 alkyl
  • R 10 is selected from aryl or C 3 -C 8 heterocycle
  • Z is O, S, NH, or NR 12 , wherein R 12 is d-C 8 alkyl;
  • R 11 is selected from H, C1-C20 alkyl, aryl, C 3 -C 8 heterocycle, -(R 13 0) m -R 14 , or - (R 13 0) m -CH(R 15 ) 2 ;
  • n is an integer ranging from
  • R 13 is C 2 -C 8 alkyl
  • R 14 is H or Ci-C 8 alkyl
  • R 15 is independently H, COOH, -(CH 2 ) n -N(R 16 ) 2 , -(CH 2 ) n -S0 3 H, or (CH 2 ) n -S0 3 -Ci-C 8 alkyl;
  • each occurrence of R is independently H, Ci-C 8 alkyl, or -(CH 2 ) n -COOH;
  • R 18 is selected from -C(R 8 ) 2 -C(R 8 ) 2 -aryl, -C(R 8 ) 2 -C(R 8 ) 2 -(C 3 -C 8 heterocycle), and
  • n is an integer ranging from 0 to 6.
  • R 3 , R 4 and R 7 are independently isopropyl or sec-butyl and R 5 is - H or methyl. In an exemplary embodiment, R 3 and R 4 are each isopropyl, R 5 is -H, and R 7 is sec-butyl.
  • R 2 and R 6 are each methyl, and R 9 is -H.
  • each occurrence of R is -OCH3.
  • R 3 and R 4 are each isopropyl
  • R 2 and R 6 are each methyl
  • R 5 is -H
  • R 7 is sec-butyl
  • each occurrence of R 8 is -OCH3
  • R 9 is -H.
  • Z is -O- or -NH-.
  • R 10 is aryl
  • R 10 is -phenyl.
  • R 11 is -H, methyl or t-butyl.
  • R 11 is -CH(R 15 ) 2 , wherein R 15 is -(CH 2 ) n -N(R 16 ) 2 , and R 16 is -Ci-C 8 alkyl or -(CH 2 ) n -COOH.
  • R 11 is -CH(R 15 ) 2 , wherein R 15 is -(CH 2 ) n - S0 3 H.
  • An exemplary auristatin embodiment of formula D E is MMAE (monomethyl auristatin E), wherein the wavy line indicates the covalent attachment to a linker of an antibody-drug conjugate:
  • An exemplary auristatin embodiment of formula Dp is MMAF (monomethyl auristatin F, a variant of auristatin E (MMAE) with a phenylalanine at the C-terminus of the drug), wherein the wavy line indicates the covalent attachment to a linker of an antibody-drug conjugate (see US 2005/0238649 and Doronina et al. (2006) Bioconjugate Chem. 17: 114-124):
  • hydrophilic groups including but not limited to, triethylene glycol esters (TEG), as shown above, can be attached to the drug moiety at R 11 .
  • TEG triethylene glycol esters
  • Exemplary embodiments of ADCs comprising an auristatin/dolastatin or derivative thereof are described in US 2005/0238649 Al and Doronina et al. (2006) Bioconjugate Chem. 17: 114-124, which is expressly incorporated herein by reference.
  • Exemplary embodiments of ADCs comprising MMAE or MMAF and various linker components have the following structures and abbreviations (wherein "Ab” is an antibody; p is the drug load (average number of drug moieties per antibody) and ranges from about 1 to about 8; "vc” is "val-cit,” i.e., a valine-citrulline dipeptide; and "S” is a sulfur atom:
  • Exemplary embodiments of ADCs comprising MMAF and various linker components further include Ab-MC-PAB-MMAF and Ab-PAB-MMAF.
  • immunoconjugates comprising MMAF attached to an antibody by a linker that is not proteolytically cleavable have been shown to possess activity comparable to immunoconjugates comprising MMAF attached to an antibody by a proteolytically cleavable linker. See, Doronina et al. (2006) Bioconjugate Chem. 17: 114-124. In such instances, drug release is believed to be effected by antibody degradation in the cell. Id.
  • peptide-based drug moieties can be prepared by forming a peptide bond between two or more amino acids and/or peptide fragments.
  • Such peptide bonds can be prepared, for example, according to the liquid phase synthesis method (see E. Schroder and K. Lubke, "The Peptides", volume 1, pp 76-136, 1965, Academic Press) that is well known in the field of peptide chemistry.
  • Auristatin/dolastatin drug moieties may be prepared according to the methods of: US 2005/0238649 Al; US Pat. No. 5635483; US Pat. No. 5780588; Pettit et al (1989) J. Am. Chem. Soc.
  • auristatin/dolastatin drug moieties of formula D F such as MMAF and derivatives thereof, may be prepared using methods described in US 2005/0238649 Al and Doronina et al. (2006) Bioconjugate Chem. 17: 114-124.
  • Auristatin/dolastatin drug moieties of formula D E such as MMAE and derivatives thereof, may be prepared using methods described in Doronina et al. (2003) Nat. Biotech. 21 :778-784.
  • Drug-linker moieties MC-MMAF, MC- MMAE, MC-vc-PAB-MMAF, and MC-vc-PAB-MMAE may be conveniently synthesized by routine methods, e.g., as described in Doronina et al. (2003) Nat. Biotech. 21 :778-784, and Patent Application Publication No. US 2005/0238649 Al, and then conjugated to an antibody of interest.
  • any of the anti-mesothelin antibodies provided herein is useful for detecting the presence of mesothelin in a biological sample.
  • the term “detecting” as used herein encompasses quantitative or qualitative detection.
  • a “biological sample” comprises, e.g., a cell or tissue (e.g., biopsy material, including cancerous or potentially cancerous pancreatic, ovarian, lung, or endometrial tissue, or mesothelioma), or serum.
  • an anti-mesothelin antibody for use in a method of diagnosis or detection is provided.
  • a method of detecting the presence of mesothelin in a biological sample comprises contacting the biological sample with an anti-mesothelin antibody as described herein under conditions permissive for binding of the anti-mesothelin antibody to mesothelin, and detecting whether a complex is formed between the anti-mesothelin antibody and mesothelin in the biological sample.
  • an anti- mesothelin antibody is used to select subjects eligible for therapy with an anti-mesothelin antibody, e.g.
  • mesothelin is a biomarker for selection of patients.
  • the biological sample is serum, e.g., wherein mesothelin that has been shed from cancer cells into the serum is detected.
  • an anti-mesothelin antibody is used in vivo to detect, e.g., by in vivo imaging, a mesothelin-positive cancer in a subject, e.g., for the purposes of diagnosing, prognosing, or staging cancer, determining the appropriate course of therapy, or monitoring response of a cancer to therapy.
  • immuno-PET immuno- positron emission tomography
  • a method for detecting a mesothelin-positive cancer in a subject comprising administering a labeled anti-mesothelin antibody to a subject having or suspected of having a mesothelin-positive cancer, and detecting the labeled anti-mesothelin antibody in the subject, wherein detection of the labeled anti-mesothelin antibody indicates a mesothelin-positive cancer in the subject.
  • the labeled anti- mesothelin antibody comprises an anti-mesothelin antibody conjugated to a positron emitter, such as 68 Ga, 18 F, 64 Cu, 86 Y, 76 Br, 89 Zr, and 124 I.
  • the positron emitter is 89 Zr.
  • a method of diagnosis or detection comprises contacting a first anti-mesothelin antibody immobilized to a substrate with a biological sample to be tested for the presence of mesothelin, exposing the substrate to a second anti-mesothelin antibody, and detecting whether the second anti-mesothelin is bound to a complex between the first anti- mesothelin antibody and mesothelin in the biological sample.
  • a substrate may be any supportive medium, e.g., glass, metal, ceramic, polymeric beads, slides, chips, and other substrates.
  • a biological sample comprises a cell or tissue (e.g., biopsy material, including cancerous or potentially cancerous pancreatic, ovarian, lung or endometrial tissue, or mesothelioma), or serum, i.e., serum in which mesothelin has been shed.
  • the first or second anti-mesothelin antibody is any of the antibodies described herein.
  • the second anti-mesothelin antibody may be 19C3 or antibodies derived from 19C3 as described herein.
  • Exemplary disorders that may be diagnosed or detected according to any of the above embodiments include mesothelin-positive cancers, such as mesothelin-positive pancreatic cancer (including pancreatic ductal adenocarcinoma), mesothelin-positive ovarian cancer (including ovarian serous adenocarcinoma), mesothelin-positive lung cancer (including non- small cell lung carcinoma (NSCLC)), mesothelioma, and mesothelin-positive endometrial cancer.
  • mesothelin-positive cancers such as mesothelin-positive pancreatic cancer (including pancreatic ductal adenocarcinoma), mesothelin-positive ovarian cancer (including ovarian serous adenocarcinoma), mesothelin-positive lung cancer (including non- small cell lung carcinoma (NSCLC)), mesothelioma, and mesothelin-positive endometrial cancer
  • a mesothelin-positive cancer is a cancer that receives an anti- mesothelin immunohistochemistry (IHC) score greater than "0," which corresponds to very weak or no staining in >90% of tumor cells, under the conditions described herein in Example J.
  • a mesothelin-positive cancer expresses mesothelin at a 1+, 2+ or 3+ level, as defined under the conditions described herein in Example J.
  • a mesothelin-positive cancer according to any of the above embodiments may be a dual-positive cancer.
  • labeled anti-mesothelin antibodies include, but are not limited to, labels or moieties that are detected directly (such as fluorescent, chromophoric, electron-dense, chemiluminescent, and radioactive labels), as well as moieties, such as enzymes or ligands, that are detected indirectly, e.g., through an enzymatic reaction or
  • Exemplary labels include, but are not limited to, the radioisotopes P, 14 C, 125 1, 3 H, and 131 I, fluorophores such as rare earth chelates or fluorescein and its derivatives, rhodamine and its derivatives, dansyl, umbelliferone, luceriferases, e.g., firefly luciferase and bacterial luciferase (U.S. Patent No.
  • luciferin 2,3-dihydrophthalazinediones
  • horseradish peroxidase HRP
  • alkaline phosphatase alkaline phosphatase
  • ⁇ -galactosidase glucoamylase
  • lysozyme saccharide oxidases, e.g., glucose oxidase, galactose oxidase, and glucose-6-phosphate dehydrogenase
  • heterocyclic oxidases such as uricase and xanthine oxidase, coupled with an enzyme that employs hydrogen peroxide to oxidize a dye precursor such as HRP
  • HRP horseradish peroxidase
  • a label is a positron emitter.
  • Positron emitters include but are not limited to 68 Ga, 18 F, 64 Cu, 86 Y, 76 Br, 89 Zr, and 124 I. In a particular embodiment, a positron emitter is 89 Zr. F. Pharmaceutical Formulations
  • compositions of an anti-mesothelin antibody or immunoconjugate as described herein are prepared by mixing such antibody or immunoconjugate having the desired degree of purity with one or more optional pharmaceutically acceptable carriers ⁇ Remington 's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), in the form of lyophilized formulations or aqueous solutions.
  • Pharmaceutically acceptable carriers are generally nontoxic to recipients at the dosages and concentrations employed, and include, but are not limited to: buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride;
  • hexamethonium chloride benzalkonium chloride; benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol;
  • polypeptides such as serum albumin, gelatin, or immunoglobulins
  • proteins such as serum albumin, gelatin, or immunoglobulins
  • hydrophilic polymers such as polyvinylpyrrolidone
  • amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine
  • monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g.
  • sHASEGP soluble neutral-active hyaluronidase glycoproteins
  • rHuPH20 HYLENEX ® , Baxter International, Inc.
  • a sHASEGP is combined with one or more additional glycosaminoglycanases such as chondroitinases.
  • Exemplary lyophilized antibody or immunoconjugate formulations are described in US Patent No. 6,267,958.
  • Aqueous antibody or immunoconjugate formulations include those described in US Patent No. 6,171,586 and WO2006/044908, the latter formulations including a histidine-acetate buffer.
  • the formulation herein may also contain more than one active ingredient as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other.
  • gemcitabine e.g., for the treatment of mesothelin-positive cancer such as mesothelin-positive pancreatic cancer (pancreatic adenocarcinoma).
  • an anti-MUC16 antibody conjugated to a cytotoxic agent e.g., for the treatment of mesothelin-positive cancer or dual-positive cancer such as mesothelin-positive ovarian cancer (ovarian serous adenocarcinoma) or dual-positive ovarian cancer.
  • Such active ingredients are suitably present in combination in amounts that are effective for the purpose intended.
  • Active ingredients may be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres,
  • microemulsions nano-particles and nanocapsules
  • macroemulsions Such techniques are disclosed in Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980).
  • sustained-release preparations may be prepared. Suitable examples of sustained- release preparations include semipermeable matrices of solid hydrophobic polymers containing the antibody or immunoconjugate, which matrices are in the form of shaped articles, e.g. films, or microcapsules.
  • the formulations to be used for in vivo administration are generally sterile. Sterility may be readily accomplished, e.g., by filtration through sterile filtration membranes. G. Therapeutic Methods and Compositions
  • anti-mesothelin antibodies or immunoconjugates may be used in methods, e.g., therapeutic methods.
  • an anti-mesothelin antibody or immunoconjugate provided herein is used in a method of inhibiting proliferation of a mesothelin-positive cell, the method comprising exposing the cell to the anti-mesothelin antibody or immunoconjugate under conditions permissive for binding of the anti-mesothelin antibody or immunoconjugate to mesothelin on the surface of the cell, thereby inhibiting the proliferation of the cell.
  • the method is an in vitro or an in vivo method.
  • the cell is a pancreatic, ovarian, lung, mesothelioma, or endometrial cell.
  • the cell is a dual- positive cell.
  • Luminescent Cell Viability Assay which is commercially available from Promega (Madison, WI). That assay determines the number of viable cells in culture based on quantitation of ATP present, which is an indication of metabolically active cells. See Crouch et al. (1993) J.
  • the assay may be conducted in 96- or 384- well format, making it amenable to automated high-throughput screening (HTS). See Cree et al. (1995) Anticancer Drugs 6:398-404.
  • the assay procedure involves adding a single reagent (CellTiter-Glo ® Reagent) directly to cultured cells. This results in cell lysis and generation of a luminescent signal produced by a luciferase reaction.
  • the luminescent signal is proportional to the amount of ATP present, which is directly proportional to the number of viable cells present in culture. Data can be recorded by luminometer or CCD camera imaging device.
  • the luminescence output is expressed as relative light units (RLU).
  • an anti-mesothelin antibody or immunoconjugate for use as a medicament is provided.
  • an anti-mesothelin antibody or immunoconjugate for use in a method of treatment is provided.
  • an anti-mesothelin antibody or immunoconjugate for use in treating mesothelin-positive cancer is provided.
  • the invention provides an anti-mesothelin antibody or immunoconjugate for use in a method of treating an individual having a mesothelin-positive cancer, the method comprising administering to the individual an effective amount of the anti-mesothelin antibody or immunoconjugate.
  • the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, e.g., as described below.
  • the invention provides for the use of an anti-mesothelin antibody or immunoconjugate in the manufacture or preparation of a medicament.
  • the medicament is for treatment of mesothelin-positive cancer.
  • the medicament is for use in a method of treating mesothelin-positive cancer, the method comprising administering to an individual having mesothelin-positive cancer an effective amount of the medicament.
  • the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, e.g., as described below.
  • the invention provides a method for treating mesothelin-positive cancer.
  • the method comprises administering to an individual having such mesothelin-positive cancer an effective amount of an anti-mesothelin antibody or
  • the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, as described below.
  • a mesothelin-positive cancer may be, e.g., mesothelin-positive pancreatic cancer (including pancreatic ductal adenocarcinoma), mesothelin-positive ovarian cancer (including ovarian serous adenocarcinoma), mesothelin- positive lung cancer (including non-small cell lung carcinoma (NSCLC)), mesothelioma, and mesothelin-positive endometrial cancer.
  • mesothelin-positive pancreatic cancer including pancreatic ductal adenocarcinoma
  • mesothelin-positive ovarian cancer including ovarian serous adenocarcinoma
  • mesothelin- positive lung cancer including non-small cell lung carcinoma (NSCLC)
  • NSCLC non-small cell lung carcinoma
  • a mesothelin-positive cancer is a cancer that receives an anti-mesothelin immunohistochemistry (IHC) score greater than "0," which corresponds to very weak or no staining in >90% of tumor cells, under the conditions described herein in Example J.
  • a mesothelin-positive cancer expresses mesothelin at a 1+, 2+ or 3+ level, as defined under the conditions described herein in Example J.
  • a mesothelin-positive cancer according to any of the above embodiments may be a dual- positive cancer.
  • An "individual” may be a human.
  • the invention provides pharmaceutical formulations comprising any of the anti-mesothelin antibodies or immunoconjugate provided herein, e.g., for use in any of the above therapeutic methods.
  • a pharmaceutical formulation comprises any of the anti-mesothelin antibodies or immunoconjugates provided herein and a pharmaceutically acceptable carrier.
  • a pharmaceutical formulation comprises any of the anti-mesothelin antibodies or immunoconjugates provided herein and at least one additional therapeutic agent, e.g., as described below.
  • Antibodies or immunoconjugates of the invention can be used either alone or in combination with other agents in a therapy.
  • an antibody or immunoconjugate of the invention may be co-administered with at least one additional therapeutic agent.
  • an additional therapeutic agent is gemcitabine.
  • an additional therapeutic agent is an anti-MUC16 antibody conjugated to a cytotoxic agent.
  • Such combination therapies noted above encompass combined administration (where two or more therapeutic agents are included in the same or separate formulations), and separate administration, in which case, administration of the antibody or immunoconjugate of the invention can occur prior to, simultaneously, and/or following, administration of the additional therapeutic agent and/or adjuvant.
  • Antibodies or immunoconjugates of the invention can also be used in combination with radiation therapy.
  • An antibody or immunoconjugate of the invention can be administered by any suitable means, including parenteral, intrapulmonary, and intranasal, and, if desired for local treatment, intralesional administration.
  • Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous
  • Dosing can be by any suitable route, e.g. by injections, such as intravenous or subcutaneous injections, depending in part on whether the administration is brief or chronic.
  • Various dosing schedules including but not limited to single or multiple administrations over various time-points, bolus administration, and pulse infusion are contemplated herein.
  • Antibodies or immunoconjugates of the invention would be formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners.
  • the antibody or immunoconjugate need not be, but is optionally formulated with one or more agents currently used to prevent or treat the disorder in question. The effective amount of such other agents depends on the amount of antibody or immunoconjugate present in the formulation, the type of disorder or treatment, and other factors discussed above.
  • an antibody or immunoconjugate of the invention when used alone or in combination with one or more other additional therapeutic agents, will depend on the type of disease to be treated, the type of antibody or immunoconjugate, the severity and course of the disease, whether the antibody or immunoconjugate is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the antibody or immunoconjugate, and the discretion of the attending physician.
  • the antibody or immunoconjugate is suitably administered to the patient at one time or over a series of treatments.
  • about 1 ⁇ g/kg to 15 mg/kg (e.g. O. lmg/kg-lOmg/kg) of antibody or immunoconjugate can be an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion.
  • One typical daily dosage might range from about 1 ⁇ g/kg to 100 mg/kg or more, depending on the factors mentioned above.
  • the treatment would generally be sustained until a desired suppression of disease symptoms occurs.
  • One exemplary dosage of the antibody or immunoconjugate would be in the range from about 0.05 mg/kg to about 10 mg/kg.
  • one or more doses of about 0.5 mg/kg, 2.0 mg/kg, 4.0 mg/kg or 10 mg/kg (or any combination thereof) may be administered to the patient.
  • Such doses may be administered intermittently, e.g. every week or every three weeks (e.g. such that the patient receives from about two to about twenty, or e.g. about six doses of the antibody).
  • An initial higher loading dose, followed by one or more lower doses may be administered.
  • other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.
  • an article of manufacture containing materials useful for the treatment, prevention and/or diagnosis of the disorders described above comprises a container and a label or package insert on or associated with the container.
  • Suitable containers include, for example, bottles, vials, syringes, IV solution bags, etc.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container holds a composition which is by itself or combined with another composition effective for treating, preventing and/or diagnosing the disorder and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • At least one active agent in the composition is an antibody or immunoconjugate of the invention.
  • the label or package insert indicates that the composition is used for treating the condition of choice.
  • the article of manufacture may comprise (a) a first container with a composition contained therein, wherein the composition comprises an antibody or immunoconjugate of the invention; and (b) a second container with a composition contained therein, wherein the composition comprises a further cytotoxic or otherwise therapeutic agent.
  • embodiment of the invention may further comprise a package insert indicating that the compositions can be used to treat a particular condition.
  • the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution or dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • the above-referenced deposited hybridoma produces the 19C3 antibody referred to herein.
  • Figure 2 is a graphic representation of human mesothelin gene expression in various tissues, which are listed on the left. The scale across the top of the graph indicates gene expression levels based on hybridization signal intensity. Dots appear both above and below the line adjacent to each listed tissue. The dots appearing above the line represent gene expression in normal tissue, and the dots appearing below the line represent gene expression in tumor and diseased tissue. Figure 2 shows increased mesothelin gene expression in certain tumor or diseased tissues relative to their normal counterparts.
  • mesothelin shows substantial overexpression in ovarian, pancreatic, endometrial and lung tumors, including adenocarcinomas and mesotheliomas.
  • Human mesothelin expression is essentially absent in normal tissues except for normal mesothelia (peritoneum, pericardium, and pleura).
  • Monoclonal antibodies against human mesothelin were generated using the following procedures. Either human MPF:mesothelin (amino acids 34-580 of SEQ ID NO:42) or human mesothelin (SEQ ID NO:43, corresponding to amino acids 296-580 of SEQ ID NO:42), each fused to an N-terminal unizyme His (HQ)-tag, was expressed in E.Coli 58F3 and purified on a Ni-NTA column (Qiagen), followed by gel filtration on a Superdex 200 column in 20mM MES pH 6.0, 6M GdnHCl as previously described (Kirchhofer et al., 2003) and dialysis into ImM HC1 for storage at -80°C.
  • mice Five Balb/c mice (Charles River Laboratories, Hollister, CA) were hyperimmunized six times with a 2 ⁇ g mixture of the two antigens in Ribi adjuvant (Ribi Immunochem Research, Inc., Hamilton, MO). The two best mice were chosen based on high antibody titers by direct ELISA and their B-cells were pooled and fused with mouse myeloma cells (X63.Ag8.653; American Type Culture Collection, Manassas, VA) using a modified protocol analogous to one previously described (Koehler and Milstein, 1975; Hongo et al., 1995).
  • supematants were harvested from hybridomas and screened for binding to both antigens (separately) by direct ELISA.
  • ELISA-positive supematants were further screened by FACS on gD-mesothelin transfected SVT2 cells (gD is an N-terminal epitope tag used as a positive control with anti-gD antibodies).
  • Positive hybridomas were subcloned twice by limiting dilution and eleven were scaled up and antibodies purified by protein A chromatography.
  • FIG. 3 shows the isolated monoclonal antibodies, along with certain properties to be described in further detail below.
  • Monoclonal antibodies 7D9 and 22A10 were humanized as described below. Residue numbers are according to Kabat et al., Sequences of proteins of immunological interest, 5th Ed., Public Health Service, National Institutes of Health, Bethesda, MD (1991). 1. Humanization of 7D9 a) Cloning of murine 7D9 variable domains
  • the variable light (VL) and variable heavy (VH) domains were amplified using RT- PCR with degenerate primers to the heavy and light chains.
  • the forward primers were specific for the N-terminal amino acid sequence of the VL and VH regions.
  • the LC and HC reverse primers were designed to anneal to a region in the constant light (CL) and constant heavy domain 1 (CHI), which are highly conserved across species.
  • the polynucleotide sequence of the inserts was determined using routine sequencing methods.
  • the 7D9 VL and VH amino acid sequences are shown in Figures 4 and 5, respectively.
  • Variants constructed during the humanization of 7D9 were assessed in the form of an IgG.
  • the VL and VH domains from murine 7D9 were aligned with the human VL kappa I (VL KI ) and human VH subgroup III (VH n i) consensus sequences.
  • Hypervariable regions from the murine 7D9 (mu7D9) antibody were engineered into VL KI and VH AT A acceptor frameworks to generate 7D9.vl .
  • the acceptor VH framework VHA T A differs from VH n i at 3 positions: R71A, N73T, and L78A (Carter et al, Proc. Natl. Acad. Sci. USA 89:4285 (1992)).
  • the direct-graft, 7D9.vl was generated by Kunkel mutagenesis using a separate oligonucleotide for each hypervariable region.
  • Three phosphorylated oligonucleotides for either heavy chain or light chain were added to 571 ng Kunkel template in 50 mM Tris pH 7.5, 10 mM MgCl 2 in a final volume of 40 ⁇ . The mixture was annealed at 90 °C for 2 min, 50 °C for 5 min and then cooled on ice.
  • 10 ⁇ annealed template was then filled in by adding 0.5 ⁇ 100 mM ATP, 0.5 ⁇ 25 mM dNTPs (25 mM each of dATP, dCTP, dGTP and dTTP), 1 ⁇ 100 mM DTT, 1 ⁇ 10X TM buffer (0.5 M Tris pH 7.5, 0.1 M MgCl 2 ), 80 U T4 ligase, and 4 U T7 polymerase in a total volume of 13.6 ⁇ for 2 hours at room temperature. 10 ⁇ of the filled in and ligated product was then transformed into XL 1 -blue cells (Stratagene). Correct clones were identified by DNA sequencing and expressed as an IgG.
  • 7D9 variants were expressed as IgG by CHO transient transfection. IgG was purified with protein G affinity chromatography. The affinity of each 7D9 IgG variant for human mesothelin was determined by surface plasmon resonance using a BIAcoreTM-2000. Biacore research grade CM5 chips were immobilized with approximately 110 RU of E. coli derived recombinant human mesothelin using the amine coupling kit from Biacore. Serial 2-fold dilutions of each 7D9 variant (0.488 to 1000 nM in PBS containing 0.05% Tween 20) were injected at a flow rate of 30 ⁇ /min. Each sample was analyzed with 5 -minute association and 3.5-minute dissociation.
  • the human acceptor framework used for humanization of 7D9 is based on the human
  • VL kappa I consensus VL KI
  • VH M human VH subgroup III consensus
  • the VL and VH domains of murine 7D9 were aligned with the human VLKI and VH m domains; hypervariable regions were identified and grafted into the human acceptor framework to generate 7D9.vl ( Figures 4 and 5).
  • the affinity of 7D9.vl is decreased ⁇ 2-fold relative to mu7D9 (formatted as a chimeric 7D9) as assessed by Biacore ( Figure 6).
  • positions 36 and 87 in the light chain and positions of 48, 67, 69, 71 , 73, 75, 76, 78 and 80 in the heavy chain were changed to residues found at these positions in mu7D9.
  • Combinations of these altered light and heavy chains with chains from 7D9.vl were transfected into CHO, expressed as IgG and purified, and assessed for binding to human mesothelin by Biacore ( Figure 6).
  • Variants 7D9.v2 and 7D9.v3 both of which contain the altered light chain, had an affinity comparable to chimeric 7D9.
  • Variant 7D9.v3 differs from 7D9.vl at 2 positions in the light chain. Neither change alone was sufficient to improve binding comparable to that of mu7D9 ( Figure 6).
  • VL and VH domains were amplified using RT-PCR with degenerate primers to the heavy chain (HC) and light chain (LC).
  • the forward primers were specific for the N-terminal amino acid sequence of the VL and VH regions.
  • the LC and HC reverse primers were designed to anneal to a region in the constant light (CL) and constant heavy domain 1 (CHI), which are highly conserved across species.
  • the polynucleotide sequence of the inserts was determined using routine sequencing methods.
  • the 22A10 VL and VH amino acid sequences are shown in Figures 7 and 8, respectively.
  • Variants constructed during the humanization of 22A10 were assessed in the form of an IgG or displayed monovalently as Fab on phage.
  • the phagemid used for this work was a monovalent Fab-g3 display vector, which consists of two open reading frames under control of a single phoA promoter. The first open reading frame consists of the stll signal sequence fused to the VL and CHI domains of the acceptor light chain, and the second consists of the stll signal sequence fused to the VH and CHI domains of the acceptor heavy chain followed by the minor phage coat protein P3.
  • VL and VH domains from murine 22A10 were aligned with the human VL kappa I (VL KI ) and human VH subgroup III (VH m ) consensus sequences.
  • Hypervariable regions from the murine 22A10 (mu22A10) antibody were engineered into VL KI and VHm acceptor frameworks to generate the 22A10 graft.
  • positions 24-34 (LI), 50-56 (L2) and 89-97 (L3) were grafted into VL KI .
  • positions 26-35 (HI), 49-65 (H2) and 95-102 (H3) were grafted into VH m ( Figures 7 and 8).
  • CDR definitions include positions defined by their sequence hypervariability (Wu, T. T. & Kabat, E. A. (1970)), their structural location (Chothia, C. & Lesk, A. M. (1987)) and their involvement in antigen-antibody contacts (MacCallum et al. J. Mol. Biol. 262: 732-745 (1996)).
  • the 22 A 10 graft was generated by Kunkel mutagenesis using a separate oligonucleotide for each hypervariable region. Three phosphorylated oligonucleotides for either heavy chain or light chain were added to 571 ng Kunkel template in 50 mM Tris pH 7.5, 10 mM MgC12 in a final volume of 40 ⁇ . The mixture was annealed at 90°C for 2 min, 50°C for 5 min and then cooled on ice.
  • 10 ⁇ annealed template was then filled in by adding 0.5 ⁇ 100 mM ATP, 0.5 ⁇ 25 mM dNTPs (25 mM each of dATP, dCTP, dGTP and dTTP), 1 ⁇ 100 mM DTT, 1 ⁇ 10X TM buffer (0.5 M Tris pH 7.5, 0.1 M MgC12), 80 U T4 ligase, and 4 U T7 polymerase in a total volume of 13.6 ⁇ for 2 hours at room temperature. 10 ⁇ of the filled in and ligated product was then transformed into XL 1 -blue cells (Stratagene). Correct clones were identified by DNA sequencing and expressed as an IgG.
  • the 22A10 graft was affinity matured using a soft randomization strategy. Sequence diversity was introduced separately into each hypervariable region such that a bias towards the murine hypervariable region sequence was maintained using a poisoned oligonucleotide synthesis strategy (Gallop et al., J Med Chem 37: 1233-51 (1994)). For each diversified position, the codon encoding the wild-type amino acid is poisoned with a 70-10-10-10 mixture of nucleotides resulting in an average 50 percent mutation rate at each position. Sequence diversity was introduced in the hypervariable regions of the 22A10-graft using Kunkel mutagenesis to generate six soft randomized phage libraries that were sorted separately. Six libraries were made each consisting of a single soft randomized hypervariable region.
  • Oligonucleotides designed to introduce diversity into each hypervariable region were phosphorylated separately in 20 ⁇ reactions containing 660 ng of oligonucleotide, 50 mM Tris pH 7.5, 10 mM MgCl 2 , 1 mM ATP, 20 mM DTT, and 5 U polynucleotide kinase for 1 h at 37°C.
  • phosphorylated oligonucleotide was added to 300 ng Kunkel template in 50 mM Tris pH 7.5, 10 mM MgCl 2 in a final volume of 10 ⁇ . The mixture was annealed at 90 °C for 2 min, 50 °C for 5 min and then cooled on ice.
  • the annealed template was then filled in by adding 0.5 ⁇ 10 mM ATP, 0.5 ⁇ 10 mM dNTPs (10 mM each of dATP, dCTP, dGTP and dTTP), 1 ⁇ 100 mM DTT, 1 ⁇ 10X TM buffer (0.5 M Tris pH 7.5, 0.1 M MgCl 2 ), 80 U T4 ligase, and 4 U T7 polymerase in a total volume of 20 ⁇ for 2 hours at room temperature.
  • Phage were harvested from the culture supernatant and suspended in PBS containing 5 % powdered milk and 0.05 % Tween 20 (PBSBT). Following addition of the phage library and a 1 hour incubation, microtiter wells were washed extensively with PBS containing 0.05 %
  • Tween 20 and bound phage were eluted by incubating the wells with 20 mM HCl, 500 mM KC1 for 30 minutes. Eluted phage were neutralized with 1 M Tris, pH 8 and amplified using XL 1 -Blue cells and M13/K07 helper phage and grown overnight at 37 °C in 2YT, 50 ⁇ g/ml carbencillin. The titers of phage eluted from a target-containing well were compared to titers of phage recovered from a non-target-containing well to assess enrichment.
  • biotinylated 293-derived human or biotinylated cynomolgus monkey mesothelin was added to phage suspended in PBS containing 5% powdered milk and 0.05 % Tween 20 (PBSBT).
  • PBSBT 0.05 % Tween 20
  • phage bound to biotinylated mesothelin were captured on a microtiter plate coated with streptavidin for 5 minutes.
  • Microtiter wells were washed extensively with PBS containing 0.05 % Tween 20 (PBST) and bound phage were eluted by incubating the wells with 20 mM HC1, 500 mM KC1 for 30 minutes.
  • Eluted phage were neutralized with 1 M Tris, pH 8 and amplified using XL1- Blue cells and M13/K07 helper phage and grown overnight at 37 °C in 2YT, 50 ⁇ g/ml carbencillin.
  • the titers of phage eluted from a target-containing well were compared to titers of phage recovered from a non-target-containing well to assess enrichment.
  • the selection stringency was gradually increased both by capturing phage that bound to decreasing concentrations of biotinylated mesothelin in solution followed by capture on neutravidin for 10 minutes (on rate selection) and by increasing the washing time and temperature to allow weak binding phage to be washed away (off rate selection) (Fuh et al, J. Mol. Biol. 340: 1073-1093 (2004)).
  • IgG variants were initially produced in 293 cells.
  • Vectors coding for VL and VH (25 ⁇ g) were transfected into 293 cells using the FUGENE system (Roche, Basel, Switzerland).
  • 500 ⁇ of FuGene was mixed with 4.5 ml of DMEM media containing no FBS and incubated at room temperature for 5 minutes.
  • Each chain (25 ⁇ g) was added to this mixture and incubated at room temperature for 20 minutes and then transferred to five T-150 flasks for transfection overnight at 37 °C in 5% C0 2 .
  • the media containing the transfection mixture was removed and replaced with 23 ml PS04 media with 0.1 ml/L trace elements (A0934) and 10 mg/L insulin (A0940).
  • Cells were incubated for an additional 5 days after which the media was harvested at 1000 rpm for 5 minutes and sterile filtered using a 0.22 ⁇ low protein-binding filter. Samples could be stored at 4°C after addition of 2.5 ml 0.1% PMSF for every 125 ml of media.
  • IgG was purified with protein G affinity chromatography.
  • the affinity of 22A10 IgG variants for human or cynomolgus monkey mesothelin was determined by surface plasmon resonance using a BIAcoreTM-2000.
  • Biacore research grade CM5 chips were immobilized with approximately 110 RU of E. coli derived recombinant human or cynomolgus monkey mesothelin using the amine coupling kit from Biacore.
  • Serial 2- fold dilutions of each 22A10 variant (0.488 to 1000 nM in PBS containing 0.05% Tween 20) were injected at a flow rate of 30 ⁇ /min. Each sample was analyzed with 5 -minute association and 3.5-minute dissociation.
  • the human acceptor framework used for humanization of 22A10 was based on the consensus human kappa I VL domain and the consensus human subgroup III VH domain.
  • the VL and VH domains of mu22A10 were aligned with the human kappa I and subgroup III domains; each complementarity determining region (CDR) was identified and grafted into the human acceptor framework to generate a CDR graft that could be expressed as an IgG or displayed as an Fab on phage ( Figures 7 and 8).
  • Humanized 22A10 variants were used to immunoprecipitate mesothelin from a cell line stably expressing mesothelin.
  • BJAB cells stably expressing gD-tagged mesothelin of different species were immunoprecipitated with the humanized 22A10 variants, as shown in Figure 9B (Gr, graft; vl (1), vl7 (17) and v83 (83)) or h7D9.v3, h5B6 anti-gD or hlgG negative control for comparison.
  • Immunoprecipitates were washed and Western blotted with murine anti-gD antibodies to detect gD-mesothelin.
  • h2210.v83 was the best of the h22A10 variants in its ability to immunoprecipitate all three species of mesothelin (cynomolgus monkey, upper;
  • the right-most lane shows 20% input lysate (without immunoprecipitation) for comparison of total expression levels.
  • Molecular weight markers (kDa) are indicated on the left.
  • mouse monoclonal antibodies 7D9 and 22A10 are referred to in the alternative as 7D9, m7D9 or mu7D9; and 22A10, m22A10 or mu22A10, respectively.
  • Humanized monoclonal antibodies 7D9.v3 and 22A10.v83 are referred to in the alternative as 7D9.v3, h7D9.v3 or hu7D9.v3; and 22A10.v83, h22A10.v83 or hu22A10.v83, respectively, unless otherwise indicated.
  • FIG. 11 shows the sequence homology between human (SEQ ID NO:43), cynomolgus monkey (SEQ ID NO:46), rat (SEQ ID NO:47) and mouse (SEQ ID NO:48) mesothelin. Shaded residues are identical between at least two species. Unshaded residues differ between at least two of the four species.
  • Figure 12 shows the results of FACS analysis of 293 cells stably transfected with gD epitope-tagged mesothelin (human, cyno, rat or mouse mesothelin); stained with 10 ⁇ g/ml h7D9.v3, h22A10.v83 or anti-gD h5B6; and detected with Alexa 647 anti-human antibody.
  • Untransfected 293 cells do not normally express mesothelin ("WT").
  • h7D9.v3 is specific for human mesothelin, while h22A10.v83 binds to human, cyno and rat mesothelin, but not mouse mesothelin.
  • Anti-gD staining verified that mouse mesothelin was indeed expressed.
  • h7D9.v3 bound to gD-tagged human mesothelin expressed on stably transfected 293, BJAB and HT1080 cell lines (all of which do not express endogenous mesothelin) with affinities of 0.2, 0.25 and 0.97 nM, respectively.
  • Kd values encompass the range seen for endogenous mesothelin in four pancreatic and two ovarian cell lines (0.41-1 nM).
  • h22A10.v83 affinities for human mesothelin expressed on the same stable cell lines were 2.7, 1.8 and 6.2 nM respectively, in accordance with its affinity for endogenous human mesothelin (-9-10 nM).
  • h22A10.v83 bound to rat mesothelin expressed on stably transfected 293 cells and BJAB cells with affinities of 7.3 nM and 2.7 nM, respectively, which is in line with the Kd of 6.2 nM observed for endogenous rat mesothelin on a normal pleural cell line, 4/4-RM4 (Aronson et al, InVitro 17: 61-70 (1981)).
  • epitope mapping of the monoclonal antibodies was performed by a standard cross-blocking ELISA.
  • Ninety-six well Nunc Immunosorp plates (Nalge Nunc, USA) were coated overnight at 4°C with 100 of 1 ⁇ g/mL human mesothelin extracellular domain in coating buffer (50 mM sodium carbonate, pH 9.5). All the following steps were performed at room temperature.
  • anti-mesothelin antibodies were also added (100 L) to a final concentration of
  • the chromogenic reaction was terminated by addition of 100 ⁇ ⁇ stop reagent (BioFX Laboratories), and the absorbance was read at 620 nm on an Ultramicroplate Reader (Biotek Instruments; Winooski, VT).
  • the maximal extent of possible binding of each of the biotinylated antibodies was determined in parallel by incubating them with mesothelin in the absence of the non-biotinylated antibodies 7D9 and 22A10.
  • anti-mesothelin antibodies indicates lack of competition for the first antibody (the maximal binding by each biotinylated antibody in the absence of the first antibody for comparison is also shown in the right group).
  • 7D9 referred to as 7D9.5.2 in Figure 14
  • 22A10 referred to as 22A10.1.2 in Figure 14
  • binds normally black bar in left group.
  • Tryptic peptide mapping experiments were performed in which h7D9.v3 was bound to immobilized human mesothelin, which was then incubated with trypsin, and the remaining antibody-protected peptides were eluted and identified by mass spectrometry. Those experiments implicated amino acids 133-183 of SEQ ID NO:43 as the h7D9.v3 binding site. To confirm this region, we took advantage of 7D9 reacting with human (construct #387 shown in Figure 15), but not mouse (construct #385) or cyno (construct #383) mesothelin to generate chimerae, which we predicted should fold better than truncation mutants.
  • the gD-tagged, GPI anchored mesothelin constructs shown in Figure 15 were transiently expressed in 293 cells and stained with 0.02 ⁇ g/ml murine 7D9, 1 ⁇ g/ml murine 22A10, or 1 ⁇ g/ml anti-gD tag (to normalize for differential expression levels). After detection with Alexa 488 anti-mouse antibody, samples were washed and analyzed by FACS, and the fluorescence intensity data were normalized to the anti-gD signal after subtraction of any background staining on wild type 293 negative control cells.
  • 7D9 binds to the human:mouse chimera #399 (having human amino acids 1-213), but not to either full length mouse mesothelin #385 or #398 (having human amino acids only from 1-131), indicating that 7D9 binds to an epitope between aa 131 and 213. Its ability to bind the cyno:human chimera #400 (having human amino acids 131-178), but not full length cyno (#383), narrowed the epitope to between amino acids 131 and 178. (Note that the relatively lower % binding seen with 7D9 than 22A10 is due to use of 5 Ox lower antibody concentration for 7D9).
  • mutant #11 from Figure 17 All mutants except mutant #11 from Figure 17 were expressed in 293 cells and subjected to FACS analysis as in Figure 16, except that 5 ⁇ g/ml humanized versions of each antibody (i.e., h7D9.v3, h22A10.v83 and h5B6 anti-gD tag (positive control)) were used, with Alexa488 anti-human antibody used for detection. Results are shown in Figure 18A, with fluorescence data shown as a percent of the anti-gD signal to normalize for expression levels. (Note: mutant #13 did not express in 293 cells and so is omitted from the dataset). h7D9.v3 bound to all mutants except #6 and #9, while h22A10.v83 bound all mutants except mutant #15 (arrows).
  • a FACS assay is performed in which binding of the test antibody to OVCAR3 cells is compared to binding of h7D9.v3 to OVCAR3 cells.
  • Suitable secondary antibodies are used to detect binding of h7D9.v3 and the test antibody to OVCAR3 cells (e.g., Alexa 647 anti-human antibody is used to detect binding of h7D9.v3).
  • Alexa 647 anti-human antibody is used to detect binding of h7D9.v3
  • Monoclonal antibodies were tested to determine whether they were capable of blocking binding of MUC 16 to mesothelin. Binding of a purified biotinylated fragment of MUC 16 (Mucl6-Bt, having three mucin repeats) to mesothelin stably expressed on A431 cells (which normally do not express mesothelin) is shown in Figure 21 ('no Ab"), left panel. Preincubation of cells with 5-fold molar ratio of 19C3, but not 7D9, inhibited the binding of MUC16-Bt to mesothelin, as detected by FACS with streptavidin-PE, as shown in Figure 21, left panel.
  • FFPE paraffin embedded
  • Figure 22 shows that 70% of pancreatic ductal adenocarcinomas were mesothelin- positive, showing staining at the 1+, 2+, or 3+ levels, with 33% showing 2+ or 3+ staining.
  • Figure 23 shows that 98% of ovarian serous adenocarcinomas were mesothelin-positive, with 74% showing staining at the 2+ or 3+ level. Additionally, all of eight tested metastases from ovarian serous adenocarcinomas were mesothelin-positive, suggesting that primary ovarian tumors do not lose mesothelin expression following metastasis.
  • Figure 24 shows that 44% of non small cell lung carcinomas (NSCLC, adenocarcinoma subtype) were mesothelin-positive, with 26%o showing staining at the 2+ or 3+ level. Additionally, three of eight (38%) tested matched metastases from mesothelin-positive primary NSCLC patient tumors retained mesothelin-positive staining.
  • NSCLC non small cell lung carcinomas
  • Mesothelin is also expressed in mesotheliomas and in endometrial cancer, as determined by IHC using the 19C3 antibody.
  • mesothelin in cynomolgous monkey was also examined. Lung pleural and heart pericardial mesothelia sections from human (formalin fixed paraffin embedded sections) and cynomolgus monkey (frozen sections) were sectioned and stained with 19C3 monoclonal antibody or 22A10 monoclonal antibody, respectively. Human mesothelia specifically stained with 19C3 ( Figure 25, left), and cynomolgus monkey mesothelia specifically stained with 22A10 ( Figure 25, right). These results demonstrate that 22A10 can recognize endogenous cynomolgus monkey mesothelin, which has a distribution similar to that in human.
  • ADCs Anti-mesothelin antibody-drug conjugates
  • the drug-linker moiety MC-vc-PAB-MMAE is otherwise referred to in these Examples and in the Figures as “vcMMAE” or "VCE.”
  • vcMMAE drug-linker moiety
  • VCE drug-linker moiety
  • h7D9.v3 -MC-vc-PAB-MMAE is referred to in these Examples and in the Figures as h7D9.v3-vcMMAE or h7D9.v3-VCE.
  • the antibodies Prior to conjugation, the antibodies were partially reduced with TCEP using standard methods in accordance with the methodology described in WO 2004/010957 A2. The partially reduced antibodies were conjugated to the drug-linker moiety using standard methods in accordance with the methodology described in Doronina et al. (2003) Nat. Biotechnol.
  • HPAC cells mesothelin-positive (2+) by IHC with 19C3
  • HBSS adenocarcinoma xenograft model.
  • Five million HPAC cells (mesothelin-positive (2+) by IHC with 19C3) in HBSS were injected subcutaneously into SCID beige mice and tumors were dosed at 1.1, 2.7, 5.5, 11, and 16.4 mg/kg h7D9.v3-vcMMAE (at 3.5 MMAE/antibody), or 5, 10 and 15 mg/kg h5B6 anti-gD-vcMMAE (with 3.3 MMAE per antibody), or with 15 mg/kg naked h7D9.v3 (no MMAE).
  • adenocarcinoma model (Oncotest, GMBH, Germany). Chunks of primary human mesothelin- positive pancreatic tumors (expressing mesothelin at 1-2+ by IHC) were implanted
  • h7D9.v3-vcMMAE The efficacy of h7D9.v3-vcMMAE was investigated using an ovarian cancer xenograft model.
  • Ten million OvCar3x2.1 cells (Mesothelin-positive (2-3+) by IHC with 19C3) were injected into the mammary fat pad of CB17 SCID beige mice, which were subsequently dosed with 1, 2.5, 5, 10 and 15 mg/kg h7D9.v3-vcMMAE (3.5 MMAE/antibody) or h5B6 anti-gD- vcMMAE (3.3 MMAE/antibody).
  • h7D9.v3-vcMMAE The efficacy of h7D9.v3-vcMMAE was investigated using a lung cancer (squamous cell carcinoma) xenograft model. Five million H226x2 cells (mesothelin-positive (3+) by IHC) were injected in a 50:50 mix of MatrigekHBSS into the flank of CB17 SCID mice. Mean tumor volumes ⁇ standard deviations are plotted in Figure 29. h7D9v3-vcMMAE (3.5
  • MMAE/antibody showed modest activity at 5mg/kg and tumor stasis at lOmg/kg, while there was no significant activity with the control anti-gD-vcMMAE conjugate (3.97
  • MMAE/antibody at either dose.
  • the right hand panel of Figure 29 shows FACS analysis and internalization of h7D9.v3 in H226x2 cells and IHC.
  • h7D9.v3-vcMMAE compared to h22A10.v83-vcMMAE was investigated.
  • mice were dosed with 0.5 or 2 mg/kg h7D9.v3-vcMMAE (in mice inoculated with BJAB-gD-human mesothelin) or h22A10.v83-vcMMAE (in mice inoculated with BJAB-gD-cynomolgous monkey mesothelin), or with anti-gD-vcMMAE at 2mg/kg used as a positive control and as a normalizer for any differences in expression between the two species of cell line.
  • Mean tumor volumes ⁇ standard deviations are plotted in Figure 30.
  • h22A10.v83-vcMMAE was similarly effective as h7D9.v3-vcMMAE in the H226x2 model (upper left panel), and only slightly less active in the OvCar3x2.1 model (upper right panel), as indicated by the faster regression of the tumors after the 6mg/kg dose.
  • OvCar3 cells which express both mesothelin and MUC16, were lysed in 1% NP40 buffer. As shown in Figure 32, left panel, lysates were immunoprecipitated with m7D9 or isotype control IgG and western blotted with an anti-MUC16 antibody (upper blot) or h7D9 (lower blot) to detect mesothelin:MUC16 complexes or total mesothelin, respectively.
  • m7D9 (20% non- immunoprecipitated input is shown in the left lane.)
  • m7D9 was able to co-immunoprecipitate MUC16 with mesothelin from OvCar3 cell lysates. That result demonstrates that MUC16 forms a complex with mesothelin in cell lines that express both mesothelin and MUC16 (i.e., "dual-positive" cell lines).
  • IP immunoprecipitate
  • the cell lines express mesothelin only (HP AC), MUC16 only (A431), neither (H520), or both (OvCar3, CAPAN-2, EKVX and OvCar429 cells).
  • the washed immunoprecipitates were Western blotted (WB) with murine anti-mesothelin antibody 2E5 (top) or murine anti-MUC16 B-domain (Ml 1 -like) antibody 1.B.823 (US Biological, Swampscott, MA; middle and bottom panels). Accordingly, the upper panel shows immunoprecipitated mesothelin from cell lines that express mesothelin, the middle panel shows immunoprecipitated MUC16 from cell lines that express MUC16, and the bottom panel shows co-immunoprecipitated mesothelin:MUC16 complexes, which are specific to cell lines expressing both proteins (dual -positive cell lines).
  • MUC16-biotin (lug/ml, or 9.2nM) was pre-bound to HT1080 cells expressing mesothelin.
  • 19C3 (5ug/ml) was added to determine if it could displace the pre-bound MUC16.
  • MUC16- biotin was detected with S APE detection reagent, and bound antibody was detected with Alexa488 anti-mouse antibody.
  • Figure 33 shows that 19C3 was indeed able to displace MUC16 and bind to mesothelin.
  • Antibody 7D9 (33nM), which binds to a region of mesothelin outside the MUC16 binding site, was used as a negative control and as expected was not able to displace the pre-bound MUC16. Additional experiments demonstrated that 19C3 also displaces MUC16 at 0. lug/ml, whereas antibody 2E5 can displace MUC16 only at >5ug/ml (data not shown).

Abstract

L'invention concerne des anticorps et des immunoconjugés anti-mésothéline ainsi que leurs modes d'utilisation.
EP11810734.1A 2010-12-20 2011-12-19 Anticorps et immunoconjugués anti-mésothéline Active EP2655418B1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP17194505.8A EP3296321B1 (fr) 2010-12-20 2011-12-19 Anticorps et immunoconjugués anti-mésothéline

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201061459962P 2010-12-20 2010-12-20
PCT/US2011/065895 WO2012087962A2 (fr) 2010-12-20 2011-12-19 Anticorps et immunoconjugués anti-mésothéline

Related Child Applications (1)

Application Number Title Priority Date Filing Date
EP17194505.8A Division EP3296321B1 (fr) 2010-12-20 2011-12-19 Anticorps et immunoconjugués anti-mésothéline

Publications (2)

Publication Number Publication Date
EP2655418A2 true EP2655418A2 (fr) 2013-10-30
EP2655418B1 EP2655418B1 (fr) 2017-10-04

Family

ID=45498116

Family Applications (2)

Application Number Title Priority Date Filing Date
EP11810734.1A Active EP2655418B1 (fr) 2010-12-20 2011-12-19 Anticorps et immunoconjugués anti-mésothéline
EP17194505.8A Active EP3296321B1 (fr) 2010-12-20 2011-12-19 Anticorps et immunoconjugués anti-mésothéline

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP17194505.8A Active EP3296321B1 (fr) 2010-12-20 2011-12-19 Anticorps et immunoconjugués anti-mésothéline

Country Status (25)

Country Link
US (3) US8911732B2 (fr)
EP (2) EP2655418B1 (fr)
JP (2) JP6253987B2 (fr)
KR (1) KR20140014116A (fr)
CN (1) CN107098972A (fr)
AR (1) AR084356A1 (fr)
AU (1) AU2011349443B2 (fr)
BR (1) BR112013014527A2 (fr)
CA (1) CA2819269A1 (fr)
CL (2) CL2013001776A1 (fr)
CO (1) CO6720984A2 (fr)
CR (1) CR20130335A (fr)
EA (2) EA201790664A1 (fr)
EC (1) ECSP13012784A (fr)
IL (1) IL250764A0 (fr)
MA (1) MA34881B1 (fr)
MX (1) MX345519B (fr)
NZ (1) NZ610976A (fr)
PE (2) PE20141114A1 (fr)
PH (1) PH12018500046A1 (fr)
SG (2) SG191294A1 (fr)
TW (2) TWI589589B (fr)
UA (2) UA113838C2 (fr)
WO (1) WO2012087962A2 (fr)
ZA (2) ZA201303765B (fr)

Families Citing this family (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107098972A (zh) 2010-12-20 2017-08-29 霍夫曼-拉罗奇有限公司 抗间皮素抗体和免疫偶联物
CN103826661B (zh) * 2011-04-21 2019-03-05 西雅图基因公司 新的结合剂-药物缀合物(adc)及其用途
WO2014004549A2 (fr) 2012-06-27 2014-01-03 Amgen Inc. Protéines de liaison anti-mésothéline
WO2015032695A1 (fr) * 2013-09-09 2015-03-12 Ventana Medical Systems, Inc. Procédé de notation pour l'expression de la protéine mésothéline
WO2015090230A1 (fr) 2013-12-19 2015-06-25 Novartis Ag Récepteurs antigéniques chimériques de la mésothéline humaine et leurs utilisations
ES2791953T3 (es) * 2014-06-06 2020-11-06 Us Health Receptores de antígeno quimérico dirigidos a mesotelina y usos de los mismos
US10626372B1 (en) 2015-01-26 2020-04-21 Fate Therapeutics, Inc. Methods and compositions for inducing hematopoietic cell differentiation
PL3298033T5 (pl) 2015-05-18 2023-10-30 TCR2 Therapeutics Inc. Kompozycje i zastosowania medyczne do reprogramowania TCR z zastosowaniem białek fuzyjnych
KR20170007181A (ko) 2015-07-10 2017-01-18 3스캔 인크. 조직학적 염색제의 공간 다중화
TWI796283B (zh) 2015-07-31 2023-03-21 德商安美基研究(慕尼黑)公司 Msln及cd3抗體構築體
TWI829617B (zh) 2015-07-31 2024-01-21 德商安美基研究(慕尼黑)公司 Flt3及cd3抗體構築體
BR112018003339A8 (pt) * 2015-08-21 2022-10-18 Carsgen Therapeutics Ltd Anticorpos antimesotelina completamente humanos e células imunoefetoras que direcionam mesotelina
KR101782487B1 (ko) 2015-09-24 2017-09-27 재단법인 목암생명과학연구소 신규 항-메소텔린 항체 및 이를 포함하는 조성물
EP3356417A1 (fr) 2015-10-02 2018-08-08 H. Hoffnabb-La Roche Ag Molécules de liaison à l'antigène activant les cellules t bispécifiques liant la mésothéline et cd3
EA039859B1 (ru) 2016-02-03 2022-03-21 Эмджен Рисерч (Мюник) Гмбх Биспецифические конструкты антител, связывающие egfrviii и cd3
GB201612520D0 (en) 2016-07-19 2016-08-31 F-Star Beta Ltd Binding molecules
JP7109789B2 (ja) 2016-08-02 2022-08-01 ティーシーアール2 セラピューティクス インク. 融合タンパク質を使用したtcrの再プログラム化のための組成物及び方法
US20190218294A1 (en) 2016-09-09 2019-07-18 Bristol-Myers Squibb Company Use of an anti-pd-1 antibody in combination with an anti-mesothelin antibody in cancer treatment
IL292551B2 (en) 2016-10-07 2023-10-01 Tcr2 Therapeutics Inc Preparations and methods for reprogramming T-cell receptors using fusion proteins
US11851491B2 (en) 2016-11-22 2023-12-26 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
CA3045466A1 (fr) 2016-12-01 2018-06-07 Regeneron Pharmaceuticals, Inc. Anticorps anti-pd-l1 radiomarques pour imagerie immuno-pet
CN110650974B (zh) 2017-02-10 2024-04-19 瑞泽恩制药公司 用于免疫-pet成像的放射性标记的抗-lag3抗体
WO2018232020A1 (fr) 2017-06-13 2018-12-20 TCR2 Therapeutics Inc. Compositions et méthodes de reprogrammation de tcr au moyen de protéines de fusion
US10730944B2 (en) 2017-07-24 2020-08-04 Regeneron Pharmaceuticals, Inc. Anti-CD8 antibodies and uses thereof
CA3086098A1 (fr) 2017-12-19 2019-06-27 F-Star Beta Limited Sequences de liaison specifiques a pd-l1 inserees dans un domaine ch3
CN110507824A (zh) * 2018-05-21 2019-11-29 荣昌生物制药(烟台)有限公司 一种抗间皮素抗体及其抗体药物缀合物
EP3807321A4 (fr) * 2018-06-18 2022-03-02 Anwita Biosciences, Inc. Constructions anti-mésothéline et utilisations associées
GB201811415D0 (en) 2018-07-12 2018-08-29 F Star Beta Ltd Anti-Mesothelin Anti bodies
US11911484B2 (en) 2018-08-02 2024-02-27 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating myotonic dystrophy
US20210308273A1 (en) 2018-08-02 2021-10-07 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating dystrophinopathies
MX2021002225A (es) 2018-08-30 2021-07-16 Tcr2 Therapeutics Inc Composiciones y metodos para la reprogramacion de tcr mediante el uso de proteinas de fusion.
BR112021025699A2 (pt) * 2019-06-19 2022-03-03 Silverback Therapeutics Inc Anticorpos anti-mesotelina e imunoconjugados dos mesmos
CA3164129A1 (fr) 2019-12-20 2021-06-24 Amgen Inc. Constructions d'anticorps multispecifique agoniste de cd40 cible par la mesotheline permettant le traitement de tumeurs solides
EP4175650A1 (fr) * 2020-07-06 2023-05-10 Kiromic BioPharma, Inc. Molécules de liaison à l'isoforme de mésothéline et molécules de récepteur pd1 chimériques, cellules les contenant et leurs utilisations
AU2021318851A1 (en) * 2020-07-23 2023-03-16 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating myotonic dystrophy
US20240024502A1 (en) * 2020-10-18 2024-01-25 Ardeagen Corporation Anti-msln binding agents, conjugates thereof and methods of using the same
CN113150128B (zh) * 2020-11-24 2023-01-31 中国农业科学院哈尔滨兽医研究所(中国动物卫生与流行病学中心哈尔滨分中心) 流感病毒np蛋白的单克隆抗体及其应用
US11771776B2 (en) 2021-07-09 2023-10-03 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating dystrophinopathies
CA3226367A1 (fr) * 2021-07-09 2023-01-12 Dyne Therapeutics, Inc. Complexes de ciblage musculaire et leurs utilisations dans le traitement de dystrophinopathies
CA3226365A1 (fr) * 2021-07-09 2023-01-12 Dyne Therapeutics, Inc. Complexes de ciblage musculaire et leurs utilisations pour le traitement de dystrophinopathies
US11633498B2 (en) 2021-07-09 2023-04-25 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating myotonic dystrophy
WO2023006391A1 (fr) 2021-07-30 2023-02-02 Ludwig-Maximilians-Universität München Anticorps anti-cd47 et utilisation associée
CN114426582B (zh) * 2022-01-07 2022-11-25 陕西脉元生物科技有限公司 一种神经特异性烯醇化酶单克隆抗体及其制备方法和应用
US11931421B2 (en) 2022-04-15 2024-03-19 Dyne Therapeutics, Inc. Muscle targeting complexes and formulations for treating myotonic dystrophy

Family Cites Families (157)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4737456A (en) 1985-05-09 1988-04-12 Syntex (U.S.A.) Inc. Reducing interference in ligand-receptor binding assays
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
US6548640B1 (en) 1986-03-27 2003-04-15 Btg International Limited Altered antibodies
IL85035A0 (en) 1987-01-08 1988-06-30 Int Genetic Eng Polynucleotide molecule,a chimeric antibody with specificity for human b cell surface antigen,a process for the preparation and methods utilizing the same
WO1988007089A1 (fr) 1987-03-18 1988-09-22 Medical Research Council Anticorps alteres
US5770701A (en) 1987-10-30 1998-06-23 American Cyanamid Company Process for preparing targeted forms of methyltrithio antitumor agents
US5606040A (en) 1987-10-30 1997-02-25 American Cyanamid Company Antitumor and antibacterial substituted disulfide derivatives prepared from compounds possessing a methyl-trithio group
AU634186B2 (en) 1988-11-11 1993-02-18 Medical Research Council Single domain ligands, receptors comprising said ligands, methods for their production, and use of said ligands and receptors
DE3920358A1 (de) 1989-06-22 1991-01-17 Behringwerke Ag Bispezifische und oligospezifische, mono- und oligovalente antikoerperkonstrukte, ihre herstellung und verwendung
CA2026147C (fr) 1989-10-25 2006-02-07 Ravi J. Chari Agents cytotoxiques comprenant des maytansinoides et leur usage therapeutique
US5208020A (en) 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
US5959177A (en) 1989-10-27 1999-09-28 The Scripps Research Institute Transgenic plants expressing assembled secretory antibodies
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
DK0554356T3 (da) 1990-10-12 1999-06-14 Us Health Monoklonalt antistof
EP0564531B1 (fr) 1990-12-03 1998-03-25 Genentech, Inc. Methode d'enrichissement pour des variantes de l'hormone de croissance avec des proprietes de liaison modifiees
US5571894A (en) 1991-02-05 1996-11-05 Ciba-Geigy Corporation Recombinant antibodies specific for a growth factor receptor
JP4124480B2 (ja) 1991-06-14 2008-07-23 ジェネンテック・インコーポレーテッド 免疫グロブリン変異体
GB9114948D0 (en) 1991-07-11 1991-08-28 Pfizer Ltd Process for preparing sertraline intermediates
WO1993006217A1 (fr) 1991-09-19 1993-04-01 Genentech, Inc. EXPRESSION DANS L'E. COLI DE FRAGMENTS D'ANTICORPS POSSEDANT AU MOINS UNE CYSTEINE PRESENTE SOUS FORME D'UN THIOL LIBRE, ET LEUR UTILISATION DANS LA PRODUCTION D'ANTICORPS BIFONCTIONNELS F(ab')¿2?
US5587458A (en) 1991-10-07 1996-12-24 Aronex Pharmaceuticals, Inc. Anti-erbB-2 antibodies, combinations thereof, and therapeutic and diagnostic uses thereof
WO1993008829A1 (fr) 1991-11-04 1993-05-13 The Regents Of The University Of California Compositions induisant la destruction de cellules infectees par l'hiv
DE69334255D1 (de) 1992-02-06 2009-02-12 Novartis Vaccines & Diagnostic Marker für Krebs und biosynthetisches Bindeprotein dafür
IL107366A (en) 1992-10-23 2003-03-12 Chugai Pharmaceutical Co Ltd Genes coding for megakaryocyte potentiator
EP0752248B1 (fr) 1992-11-13 2000-09-27 Idec Pharmaceuticals Corporation Application thérapeutique d'anticorps chimériques et radio-marqués contre l'antigène à differentiation restreinte des lymphocytes B humains pour le traitement du lymphome des cellules B
US5635483A (en) 1992-12-03 1997-06-03 Arizona Board Of Regents Acting On Behalf Of Arizona State University Tumor inhibiting tetrapeptide bearing modified phenethyl amides
US5780588A (en) 1993-01-26 1998-07-14 Arizona Board Of Regents Elucidation and synthesis of selected pentapeptides
EP0714409A1 (fr) 1993-06-16 1996-06-05 Celltech Therapeutics Limited Anticorps
US5773001A (en) 1994-06-03 1998-06-30 American Cyanamid Company Conjugates of methyltrithio antitumor agents and intermediates for their synthesis
US5789199A (en) 1994-11-03 1998-08-04 Genentech, Inc. Process for bacterial production of polypeptides
US5663149A (en) 1994-12-13 1997-09-02 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory pentapeptide heterocyclic and halophenyl amides
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
US5840523A (en) 1995-03-01 1998-11-24 Genetech, Inc. Methods and compositions for secretion of heterologous polypeptides
US5869046A (en) 1995-04-14 1999-02-09 Genentech, Inc. Altered polypeptides with increased half-life
US5712374A (en) 1995-06-07 1998-01-27 American Cyanamid Company Method for the preparation of substantiallly monomeric calicheamicin derivative/carrier conjugates
US5714586A (en) 1995-06-07 1998-02-03 American Cyanamid Company Methods for the preparation of monomeric calicheamicin derivative/carrier conjugates
US6267958B1 (en) 1995-07-27 2001-07-31 Genentech, Inc. Protein formulation
US7375183B1 (en) 1996-01-05 2008-05-20 The United States Of America As Represented By The Department Of Health And Human Services Mesothelin, immunogenic peptides derived therefrom, and compositions comprising mesothelin, or immunogenic peptides thereof
EP0871492B1 (fr) 1996-01-05 2003-11-26 THE GOVERNMENT OF THE UNITED STATES OF AMERICA, as represented by the Secretary, Department of Health and Human Services Antigene de mesothelium, procedes et kits de ciblage de celui-ci
GB9603256D0 (en) 1996-02-16 1996-04-17 Wellcome Found Antibodies
US6171586B1 (en) 1997-06-13 2001-01-09 Genentech, Inc. Antibody formulation
CA2293829C (fr) 1997-06-24 2011-06-14 Genentech, Inc. Procedes et compositions concernant des glycoproteines galactosylees
US6040498A (en) 1998-08-11 2000-03-21 North Caroline State University Genetically engineered duckweed
US6602677B1 (en) 1997-09-19 2003-08-05 Promega Corporation Thermostable luciferases and methods of production
AU759779B2 (en) 1997-10-31 2003-05-01 Genentech Inc. Methods and compositions comprising glycoprotein glycoforms
US6610833B1 (en) 1997-11-24 2003-08-26 The Institute For Human Genetics And Biochemistry Monoclonal human natural antibodies
US6809184B1 (en) 1997-12-01 2004-10-26 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Antibodies, including FV molecules, and immunoconjugates having high binding affinity for mesothelin and methods for their use
AU760120B2 (en) 1997-12-01 2003-05-08 Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services, The Antibodies, including Fv molecules, and immunoconjugates having high binding affinity for mesothelin and methods for their use
BR9813365A (pt) 1997-12-05 2004-06-15 Scripps Research Inst Método para produção e humanização de um anticorpo monoclonal de rato
DK1068241T3 (da) 1998-04-02 2008-02-04 Genentech Inc Antistofvarianter og fragmenter deraf
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
PT1071700E (pt) 1998-04-20 2010-04-23 Glycart Biotechnology Ag Modificação por glicosilação de anticorpos para melhorar a citotoxicidade celular dependente de anticorpos
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
ES2694002T3 (es) 1999-01-15 2018-12-17 Genentech, Inc. Polipéptido que comprende una región Fc de IgG1 humana variante
US7745159B2 (en) 1999-02-26 2010-06-29 Nathalie B Scholler Methods and compositions for diagnosing carcinomas
US6770445B1 (en) 1999-02-26 2004-08-03 Pacific Northwest Research Institute Methods and compositions for diagnosing carcinomas
PT1914244E (pt) 1999-04-09 2013-07-26 Kyowa Hakko Kirin Co Ltd Processo para regular a actividade de moléculas funcionais sob o ponto de vista imunológico
DE60039510D1 (de) 1999-05-27 2008-08-28 Us Gov Health & Human Serv Immunokonjugate mit hoher bindungsaffinität
US20110104675A1 (en) 1999-08-09 2011-05-05 Pacific Northwest Research Institute Methods and Compositions for Diagnosing Carcinomas
WO2001025454A2 (fr) 1999-10-04 2001-04-12 Medicago Inc. Procede de regulation de la transcription de genes etrangers
US7125978B1 (en) 1999-10-04 2006-10-24 Medicago Inc. Promoter for regulating expression of foreign genes
JP4668498B2 (ja) 1999-10-19 2011-04-13 協和発酵キリン株式会社 ポリペプチドの製造方法
CA2393869A1 (fr) 1999-12-15 2001-06-21 Genetech,Inc. Balayage aveugle, procede combinatoire permettant la representation d'epitopes de proteines fonctionnelles
WO2001049698A1 (fr) 1999-12-29 2001-07-12 Immunogen, Inc. Agents cytotoxiques comprenant des doxorubicines et des daunorubicines modifiees et utilisation therapeutique de ceux-ci
NZ521540A (en) 2000-04-11 2004-09-24 Genentech Inc Multivalent antibodies and uses therefor
US20020132237A1 (en) 2000-05-26 2002-09-19 Corixa Corporation Compositions and methods for the therapy and diagnosis of ovarian cancer
US7064191B2 (en) 2000-10-06 2006-06-20 Kyowa Hakko Kogyo Co., Ltd. Process for purifying antibody
US6946292B2 (en) 2000-10-06 2005-09-20 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions with increased antibody dependent cytotoxic activity
CA2785941C (fr) 2000-10-06 2017-01-10 Kyowa Hakko Kirin Co., Ltd. Cellules produisant des compositions d'anticorps
US6596541B2 (en) 2000-10-31 2003-07-22 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
WO2002040545A2 (fr) 2000-11-17 2002-05-23 The Government Of The United States, As Represented By The Secretary Of The Department Of Health And Human Services Reduction de la toxicite animale non specifique des immunotoxines par mutation des regions d'infrastructure de fv pour reduire le point isoelectrique
ATE378403T1 (de) 2000-11-30 2007-11-15 Medarex Inc Transchromosomale transgen-nagetiere zur herstellung von humänen antikörpern
US20030087250A1 (en) 2001-03-14 2003-05-08 Millennium Pharmaceuticals, Inc. Nucleic acid molecules and proteins for the identification, assessment, prevention, and therapy of ovarian cancer
US6884869B2 (en) 2001-04-30 2005-04-26 Seattle Genetics, Inc. Pentapeptide compounds and uses related thereto
US7125663B2 (en) 2001-06-13 2006-10-24 Millenium Pharmaceuticals, Inc. Genes, compositions, kits and methods for identification, assessment, prevention, and therapy of cervical cancer
AU2002347428A1 (en) 2001-06-18 2003-01-02 Eos Biotechnology Inc. Methods of diagnosis of ovarian cancer, compositions and methods of screening for modulators of ovarian cancer
US7803915B2 (en) 2001-06-20 2010-09-28 Genentech, Inc. Antibody compositions for the diagnosis and treatment of tumor
US20050107595A1 (en) 2001-06-20 2005-05-19 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
CN1555411A (zh) 2001-08-03 2004-12-15 ���迨�����\���ɷݹ�˾ 抗体-依赖性细胞毒性增大的抗体糖基化变体
CA2456369A1 (fr) 2001-08-08 2003-02-20 Incyte Genomics, Inc. Proteines associees a la croissance, la differentiation et la mort cellulaires
EP1443961B1 (fr) 2001-10-25 2009-05-06 Genentech, Inc. Compositions de glycoproteine
CA2467242A1 (fr) 2001-11-20 2003-05-30 Seattle Genetics, Inc. Traitement des troubles immunologiques au moyen des anticorps anti-cd30
US20040093621A1 (en) 2001-12-25 2004-05-13 Kyowa Hakko Kogyo Co., Ltd Antibody composition which specifically binds to CD20
US7691568B2 (en) 2002-04-09 2010-04-06 Kyowa Hakko Kirin Co., Ltd Antibody composition-containing medicament
JPWO2003085107A1 (ja) 2002-04-09 2005-08-11 協和醗酵工業株式会社 ゲノムが改変された細胞
JP4628679B2 (ja) 2002-04-09 2011-02-09 協和発酵キリン株式会社 Gdp−フコースの輸送に関与する蛋白質の活性が低下または欠失した細胞
CA2481925A1 (fr) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. Agent therapeutique pour les patients atteints du fc.gamma.riiia humain
EP1498490A4 (fr) 2002-04-09 2006-11-29 Kyowa Hakko Kogyo Kk Procede de production de composition anticorps
AU2003236018A1 (en) 2002-04-09 2003-10-20 Kyowa Hakko Kirin Co., Ltd. METHOD OF ENHANCING ACTIVITY OF ANTIBODY COMPOSITION OF BINDING TO FcGamma RECEPTOR IIIa
US20050276812A1 (en) 2004-06-01 2005-12-15 Genentech, Inc. Antibody-drug conjugates and methods
JP4753578B2 (ja) 2002-06-03 2011-08-24 ジェネンテック, インコーポレイテッド 合成抗体ファージライブラリー
WO2003101283A2 (fr) 2002-06-04 2003-12-11 Incyte Corporation Marqueurs diagnostiques du cancer du poumon
EP1575500A4 (fr) 2002-07-12 2007-01-03 Univ Johns Hopkins Vaccins a la mesotheline et systemes de modele
EP2357006B1 (fr) 2002-07-31 2015-09-16 Seattle Genetics, Inc. Conjugués de médicaments et leur utilisation pour traiter le cancer, maladie auto-immune ou maladie infectieuse
US7361740B2 (en) 2002-10-15 2008-04-22 Pdl Biopharma, Inc. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
EP1572744B1 (fr) 2002-12-16 2010-06-09 Genentech, Inc. Variants d'immunoglobuline et utilisations
WO2004065416A2 (fr) 2003-01-16 2004-08-05 Genentech, Inc. Banques de phages anticorps synthetiques
US7871607B2 (en) 2003-03-05 2011-01-18 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminoglycanases
US20060104968A1 (en) 2003-03-05 2006-05-18 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminogly ycanases
US20040180387A1 (en) 2003-03-13 2004-09-16 Fujirebio Diagnostics, Inc. Detection of urinary mesothelin-/megakaryocyte potentiating factor-related peptides for assessment of ovarian cancer
EP1651675A1 (fr) 2003-08-05 2006-05-03 Morphotek, Inc. Molecule a surface cellulaire variante liee au cancer
WO2005035586A1 (fr) 2003-10-08 2005-04-21 Kyowa Hakko Kogyo Co., Ltd. Composition proteique hybride
EP1705251A4 (fr) 2003-10-09 2009-10-28 Kyowa Hakko Kirin Co Ltd Procede permettant de produire une composition d'anticorps par inhibition par l'arn de la fonction de $g(a)1,6-fucosyltransferase
RS55723B1 (sr) 2003-11-05 2017-07-31 Roche Glycart Ag Molekuli koji se vezuju za antigen sa povećanim afinitetom vezivanja za fc receptor i efektornom funkcijom
BR122018071968B8 (pt) 2003-11-06 2021-07-27 Seattle Genetics Inc conjugado de anticorpo-droga, composição farmacêutica, artigo de manufatura e uso de um conjugado de anticorpo-droga
WO2005053742A1 (fr) 2003-12-04 2005-06-16 Kyowa Hakko Kogyo Co., Ltd. Medicament contenant une composition a base d'anticorps
WO2005072263A2 (fr) 2004-01-21 2005-08-11 Fujirebio America, Inc. Detection de peptides relatifs au facteur de potentialisation des mesotheline-/megacaryocytes urinaires aux fins d'evaluation de mesotheliome
EP2444805A3 (fr) 2004-01-21 2012-06-20 Fujirebio America, Inc. Détection de peptides relatifs au facteur de potentialisation des mésothélines-/mégacaryocytes dans le fluide péritonéal à des fins d'évaluation du péritoine et de la cavité péritonéale
EP1718667B1 (fr) 2004-02-23 2013-01-09 Genentech, Inc. Liants et conjugues heterocycliques auto-immolateurs
RU2386638C2 (ru) 2004-03-31 2010-04-20 Дженентек, Инк. Гуманизированные анти-тфр-бета-антитела
US7662936B2 (en) 2004-04-07 2010-02-16 Genentech, Inc. Mass spectrometry of antibody conjugates
US7785903B2 (en) 2004-04-09 2010-08-31 Genentech, Inc. Variable domain library and uses
CA2885854C (fr) 2004-04-13 2017-02-21 F. Hoffmann-La Roche Ag Anticorps anti-p-selectine
EP1782070B1 (fr) 2004-06-17 2010-09-08 MannKind Corporation Combinaisons d'antigenes associes a une tumeur pour le diagnostic de differents types de cancer
TWI380996B (zh) 2004-09-17 2013-01-01 Hoffmann La Roche 抗ox40l抗體
US20100111856A1 (en) 2004-09-23 2010-05-06 Herman Gill Zirconium-radiolabeled, cysteine engineered antibody conjugates
NZ580115A (en) 2004-09-23 2010-10-29 Genentech Inc Cysteine engineered antibody light chains and conjugates
JO3000B1 (ar) 2004-10-20 2016-09-05 Genentech Inc مركبات أجسام مضادة .
US20070134243A1 (en) 2004-12-01 2007-06-14 Gazzard Lewis J Antibody drug conjugates and methods
US7947839B2 (en) 2004-12-01 2011-05-24 Genentech, Inc. Heterocyclic-substituted bis-1,8 naphthalimide compounds, antibody drug conjugates, and methods of use
JP4993645B2 (ja) 2004-12-01 2012-08-08 ジェネンテック, インコーポレイテッド 抗体薬剤結合体および方法
WO2006065867A2 (fr) 2004-12-14 2006-06-22 Enzon Pharmaceuticals Inc. Immunotoxine exotoxique de pseudomonas a liaison polymere
JPWO2006068204A1 (ja) 2004-12-21 2008-06-12 旭硝子株式会社 透明導電膜付き基板とそのパターニング方法
US7592426B2 (en) * 2005-03-10 2009-09-22 Morphotek, Inc. Anti-mesothelin antibodies
CA2607305A1 (fr) 2005-05-12 2006-11-23 The Government Of The United States Of America As Represented By The Sec Retary Of The Department Of Health And Human Services Anticorps diriges contre la mesotheline utiles pour des dosages immunologiques
WO2007100385A2 (fr) 2005-10-31 2007-09-07 Genentech, Inc. Conjugués anticorps-médicaments à base de depsipeptide macrocyclique et méthodes associées
WO2007056441A2 (fr) 2005-11-07 2007-05-18 Genentech, Inc. Polypeptides de liaison comprenant des sequences diversifiees et des sequences consensus hypervariables vh/vl
WO2007064919A2 (fr) 2005-12-02 2007-06-07 Genentech, Inc. Polypeptides de liaison avec des sequences de diversite limitees
WO2007112047A2 (fr) 2006-03-24 2007-10-04 Enzon Pharmaceuticals, Inc. Formulations contenant des immunotoxines ciblées sur l'antigène de cellules mésothéliennes
AU2007249408A1 (en) 2006-05-09 2007-11-22 Genentech, Inc. Binding polypeptides with optimized scaffolds
CA2652538C (fr) 2006-05-19 2016-06-28 Teva Pharmaceutical Industries, Ltd. Proteines de fusion, leurs utilisations et leurs procedes de production
US10118970B2 (en) 2006-08-30 2018-11-06 Genentech, Inc. Multispecific antibodies
CN101563454A (zh) 2006-12-08 2009-10-21 株式会社免疫生物研究所 间皮瘤诊断剂、间皮瘤诊断试剂盒和间皮瘤诊断方法
AU2007332473B2 (en) 2006-12-14 2012-09-27 Forerunner Pharma Research Co., Ltd. Anti-Claudin-3 monoclonal antibody, and treatment and diagnosis of cancer using the same
CA2650077A1 (fr) 2006-12-19 2008-02-17 Ekaterina Dadachova Radioimmunotherapie et imagerie de cellules tumorales exprimant des antigenes viraux
US20080226635A1 (en) 2006-12-22 2008-09-18 Hans Koll Antibodies against insulin-like growth factor I receptor and uses thereof
EP2144628B1 (fr) * 2007-05-08 2012-10-17 Genentech, Inc. Anticorps anti-muc16 produits avec de la cystéine et conjugués anticorps-médicament
CN100592373C (zh) 2007-05-25 2010-02-24 群康科技(深圳)有限公司 液晶显示面板驱动装置及其驱动方法
CA2700860C (fr) 2007-10-01 2016-07-19 Jonathan A. Terrett Anticorps humains qui se lient a la mesotheline, et utilisations de ceux-ci
PL2215121T3 (pl) * 2007-11-26 2016-07-29 Bayer Ip Gmbh Przeciwciała przeciwko mezotelinie i ich zastosowania
WO2009089004A1 (fr) 2008-01-07 2009-07-16 Amgen Inc. Méthode de fabrication de molécules hétérodimères fc d'anticorps utilisant les effets de conduite électrostatique
US8742076B2 (en) 2008-02-01 2014-06-03 Genentech, Inc. Nemorubicin metabolite and analog reagents, antibody-drug conjugates and methods
EP2257572A1 (fr) 2008-03-27 2010-12-08 The Government of the United States of America as represented by The Secretary of the Department of Health and Human Services Anticorps monoclonaux anti-mésothéline humaine
JP6067222B2 (ja) 2008-07-15 2017-01-25 ジェネンテック, インコーポレイテッド アントラサイクリン誘導体コンジュゲート、その調製方法及び抗腫瘍化合物としてのその用途
SG173812A1 (en) 2009-02-27 2011-09-29 Genentech Inc Methods and compositions for protein labelling
CA2756393C (fr) * 2009-03-24 2017-06-20 The Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services Anticorps anti-mesotheline
AR076284A1 (es) * 2009-04-29 2011-06-01 Bayer Schering Pharma Ag Inmunoconjugados de antimesotelina y usos de los mismos
PE20130342A1 (es) 2010-04-15 2013-04-20 Spirogen Sarl Pirrolobenzodiacepinas y conjugados de las mismas
CN114246952A (zh) 2010-06-08 2022-03-29 基因泰克公司 半胱氨酸改造的抗体和偶联物
WO2012055019A1 (fr) 2010-10-29 2012-05-03 Urban Surface Solutions Inc. Dispositif de chauffage à jets
JP5889912B2 (ja) 2010-11-17 2016-03-22 ジェネンテック, インコーポレイテッド アラニニルメイタンシノール抗体コンジュゲート
CN107098972A (zh) 2010-12-20 2017-08-29 霍夫曼-拉罗奇有限公司 抗间皮素抗体和免疫偶联物
CA2825064C (fr) 2011-02-04 2022-08-30 Genentech, Inc. Variantes genetiques de fc et leurs procedes de production
JP5987053B2 (ja) 2011-05-12 2016-09-06 ジェネンテック, インコーポレイテッド フレームワークシグネチャーペプチドを用いて動物サンプルにおける治療抗体を検出するための多重反応モニタリングlc−ms/ms法
PT2750713E (pt) 2011-10-14 2016-01-20 Genentech Inc Pirrolobenzodiazepinas e conjugados das mesmas

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2012087962A2 *

Also Published As

Publication number Publication date
JP6311042B2 (ja) 2018-04-11
IL250764A0 (en) 2017-04-30
SG10201510518TA (en) 2016-01-28
WO2012087962A2 (fr) 2012-06-28
TWI477513B (zh) 2015-03-21
EA201790664A1 (ru) 2017-07-31
EP2655418B1 (fr) 2017-10-04
ECSP13012784A (es) 2013-09-30
EP3296321B1 (fr) 2020-07-15
MX345519B (es) 2017-02-01
US20150065388A1 (en) 2015-03-05
ZA201406196B (en) 2016-09-28
CR20130335A (es) 2013-08-12
UA113838C2 (xx) 2017-03-27
BR112013014527A2 (pt) 2017-03-07
JP2017125022A (ja) 2017-07-20
PH12018500046A1 (en) 2019-04-08
EA201390933A1 (ru) 2013-12-30
AU2011349443A1 (en) 2013-05-09
CN103347897A (zh) 2013-10-09
TWI589589B (zh) 2017-07-01
WO2012087962A3 (fr) 2012-09-13
CN107098972A (zh) 2017-08-29
PE20141114A1 (es) 2014-09-15
AR084356A1 (es) 2013-05-08
UA115641C2 (uk) 2017-11-27
ZA201303765B (en) 2014-11-26
EP3296321A1 (fr) 2018-03-21
AU2011349443B2 (en) 2015-12-24
SG191294A1 (en) 2013-07-31
NZ610976A (en) 2015-07-31
US10022452B2 (en) 2018-07-17
PE20170917A1 (es) 2017-07-12
JP6253987B2 (ja) 2017-12-27
CL2013001776A1 (es) 2014-03-07
KR20140014116A (ko) 2014-02-05
MX2013006716A (es) 2013-09-26
US9719996B2 (en) 2017-08-01
EA028744B1 (ru) 2017-12-29
US20120225013A1 (en) 2012-09-06
CO6720984A2 (es) 2013-07-31
JP2014509835A (ja) 2014-04-24
CA2819269A1 (fr) 2012-06-28
US20170281792A1 (en) 2017-10-05
TW201302788A (zh) 2013-01-16
CL2017000772A1 (es) 2017-11-03
TW201536807A (zh) 2015-10-01
US8911732B2 (en) 2014-12-16
MA34881B1 (fr) 2014-02-01

Similar Documents

Publication Publication Date Title
US10022452B2 (en) Anti-mesothelin antibodies and immunoconjugates
US20230134580A1 (en) Anti-tigit antibodies and methods of use
US11702479B2 (en) Anti-jagged antibodies and methods of use
US20230037911A1 (en) Anti-tigit antibodies, multispecific antibodies comprising the same, and methods of using the same
US20230060388A1 (en) Anti-tigit antibodies, multispecific antibodies comprising the same, and methods of using the same
CA2850034A1 (fr) Associations therapeutiques et methodes de traitement du melanome
WO2022255440A1 (fr) Anticorps anti-ddr2 et leurs utilisations

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20130722

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

DAX Request for extension of the european patent (deleted)
17Q First examination report despatched

Effective date: 20140708

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

RIC1 Information provided on ipc code assigned before grant

Ipc: A61K 39/395 20060101ALI20170322BHEP

Ipc: A61P 35/00 20060101ALI20170322BHEP

Ipc: C07K 16/30 20060101AFI20170322BHEP

INTG Intention to grant announced

Effective date: 20170419

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 933920

Country of ref document: AT

Kind code of ref document: T

Effective date: 20171015

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 7

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602011042158

Country of ref document: DE

REG Reference to a national code

Ref country code: NL

Ref legal event code: MP

Effective date: 20171004

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 933920

Country of ref document: AT

Kind code of ref document: T

Effective date: 20171004

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180104

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180105

Ref country code: RS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180204

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180104

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602011042158

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

Ref country code: IT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

26N No opposition filed

Effective date: 20180705

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20171219

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20171219

REG Reference to a national code

Ref country code: BE

Ref legal event code: MM

Effective date: 20171231

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20171219

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20171231

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20111219

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20171004

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CH

Payment date: 20230101

Year of fee payment: 12

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20231124

Year of fee payment: 13

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FR

Payment date: 20231122

Year of fee payment: 13

Ref country code: DE

Payment date: 20231121

Year of fee payment: 13

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CH

Payment date: 20240101

Year of fee payment: 13